101
|
Blanke N, Go V, Rosene DL, Bigio IJ. Quantitative birefringence microscopy for imaging the structural integrity of CNS myelin following circumscribed cortical injury in the rhesus monkey. NEUROPHOTONICS 2021; 8:015010. [PMID: 33763502 PMCID: PMC7984970 DOI: 10.1117/1.nph.8.1.015010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 03/04/2021] [Indexed: 05/12/2023]
Abstract
Significance: Myelin breakdown is likely a key factor in the loss of cognitive and motor function associated with many neurodegenerative diseases. Aim: New methods for imaging myelin structure are needed to characterize and quantify the degradation of myelin in standard whole-brain sections of nonhuman primates and in human brain. Approach: Quantitative birefringence microscopy (qBRM) is a label-free technique for rapid histopathological assessment of myelin structural breakdown following cortical injury in rhesus monkeys. Results: We validate birefringence microscopy for structural imaging of myelin in rhesus monkey brain sections, and we demonstrate the power of qBRM by characterizing the breakdown of myelin following cortical injury, as a model of stroke, in the motor cortex. Conclusions: Birefringence microscopy is a valuable tool for histopathology of myelin and for quantitative assessment of myelin structure. Compared to conventional methods, this label-free technique is sensitive to subtle changes in myelin structure, is fast, and enables more quantitative assessment, without the variability inherent in labeling procedures such as immunohistochemistry.
Collapse
Affiliation(s)
- Nathan Blanke
- Boston University, Neurophotonics Center, Department of Biomedical Engineering, Boston, Massachusetts, United States
| | - Veronica Go
- Boston University School of Medicine, Department of Anatomy and Neurobiology, Boston, Massachusetts, United States
| | - Douglas L. Rosene
- Boston University School of Medicine, Department of Anatomy and Neurobiology, Boston, Massachusetts, United States
| | - Irving J. Bigio
- Boston University, Neurophotonics Center, Department of Biomedical Engineering, Boston, Massachusetts, United States
- Boston University, Department of Electrical and Computer Engineering, Boston, Massachusetts, United States
| |
Collapse
|
102
|
Shaw JC, Crombie GK, Palliser HK, Hirst JJ. Impaired Oligodendrocyte Development Following Preterm Birth: Promoting GABAergic Action to Improve Outcomes. Front Pediatr 2021; 9:618052. [PMID: 33634057 PMCID: PMC7901941 DOI: 10.3389/fped.2021.618052] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 01/12/2021] [Indexed: 11/21/2022] Open
Abstract
Preterm birth is associated with poor long-term neurodevelopmental and behavioral outcomes, even in the absence of obvious brain injury at the time of birth. In particular, behavioral disorders characterized by inattention, social difficulties and anxiety are common among children and adolescents who were born moderately to late preterm (32-37 weeks' gestation). Diffuse deficits in white matter microstructure are thought to play a role in these poor outcomes with evidence suggesting that a failure of oligodendrocytes to mature and myelinate axons is responsible. However, there remains a major knowledge gap over the mechanisms by which preterm birth interrupts normal oligodendrocyte development. In utero neurodevelopment occurs in an inhibitory-dominant environment due to the action of placentally derived neurosteroids on the GABAA receptor, thus promoting GABAergic inhibitory activity and maintaining the fetal behavioral state. Following preterm birth, and the subsequent premature exposure to the ex utero environment, this action of neurosteroids on GABAA receptors is greatly reduced. Coinciding with a reduction in GABAergic inhibition, the preterm neonatal brain is also exposed to ex utero environmental insults such as periods of hypoxia and excessive glucocorticoid concentrations. Together, these insults may increase levels of the excitatory neurotransmitter glutamate in the developing brain and result in a shift in the balance of inhibitory: excitatory activity toward excitatory. This review will outline the normal development of oligodendrocytes, how it is disrupted under excitation-dominated conditions and highlight how shifting the balance back toward an inhibitory-dominated environment may improve outcomes.
Collapse
Affiliation(s)
- Julia C Shaw
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, NSW, Australia.,Mothers and Babies Research Centre, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Gabrielle K Crombie
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, NSW, Australia.,Mothers and Babies Research Centre, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Hannah K Palliser
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, NSW, Australia.,Mothers and Babies Research Centre, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Jonathan J Hirst
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, NSW, Australia.,Mothers and Babies Research Centre, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| |
Collapse
|
103
|
Al-Griw MA, Salter MG, Wood IC. Inhibition of ionotropic GluR signaling preserves oligodendrocyte lineage and myelination in an ex vivo rat model of white matter ischemic injury. Acta Neurobiol Exp (Wars) 2021; 81:233-248. [PMID: 34672294 DOI: 10.21307/ane-2021-022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Preterm infants have a high risk of neonatal white matter injury (WMI). WMI leads to reduced myelination, inflammation, and clinical neurodevelopmental deficits for which there are no effective treatments. Ionotropic glutamate receptor (iGluR) induced excitotoxicity contributes to oligodendrocyte (OL) lineage cell loss and demyelination in brain models of neonatal and adult WMI. Here, we hypothesized that simulated ischemia (oxygen‑glucose deprivation) damages white matter via activation of iGluR signaling, and that iGluR inhibition shortly after WMI could mitigate OL loss, enhance myelination, and suppress inflammation in an ex vivo cerebellar slice model of developing WMI. Inhibition of iGluR signaling by a combined block of AMPA and NMDA receptors, shortly after simulated ischemia, restored myelination, reduced apoptotic OLs, and enhanced OL precursor cell proliferation and maturation as well as upregulated expression of transcription factors regulating OL development and remyelination. Our findings demonstrate that iGluR inhibition post‑injury alleviates OL lineage cell loss and inflammation and promotes myelination upon developing WMI. The findings may help to develop therapeutic interventions for the WMI treatment.
Collapse
Affiliation(s)
- Mohamed A Al-Griw
- Department of Histology and Genetics, Faculty of Medicine, University of Tripoli, Tripoli, Libya;
| | | | - Ian C Wood
- Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
104
|
Galinsky R, van de Looij Y, Mitchell N, Dean JM, Dhillon SK, Yamaguchi K, Lear CA, Wassink G, Davidson JO, Nott F, Zahra VA, Kelly SB, King VJ, Sizonenko SV, Bennet L, Gunn AJ. Magnetic Resonance Imaging Correlates of White Matter Gliosis and Injury in Preterm Fetal Sheep Exposed to Progressive Systemic Inflammation. Int J Mol Sci 2020; 21:ijms21238891. [PMID: 33255257 PMCID: PMC7727662 DOI: 10.3390/ijms21238891] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/17/2020] [Accepted: 11/19/2020] [Indexed: 12/17/2022] Open
Abstract
Progressive fetal infection/inflammation is strongly associated with neural injury after preterm birth. We aimed to test the hypotheses that progressively developing fetal inflammation leads to neuroinflammation and impaired white matter development and that the histopathological changes can be detected using high-field diffusion tensor magnetic resonance imaging (MRI). Chronically instrumented preterm fetal sheep at 0.7 of gestation were randomly assigned to receive intravenous saline (control; n = 6) or a progressive infusion of lipopolysaccharide (LPS, 200 ng intravenous over 24 h then doubled every 24 h for 5 days to induce fetal inflammation, n = 7). Sheep were killed 10 days after starting the infusions, for histology and high-field diffusion tensor MRI. Progressive LPS infusion was associated with increased circulating interleukin (IL)-6 concentrations and moderate increases in carotid artery perfusion and the frequency of electroencephalogram (EEG) activity (p < 0.05 vs. control). In the periventricular white matter, fractional anisotropy (FA) was increased, and orientation dispersion index (ODI) was reduced (p < 0.05 vs. control for both). Histologically, in the same brain region, LPS infusion increased microglial activation and astrocyte numbers and reduced the total number of oligodendrocytes with no change in myelination or numbers of immature/mature oligodendrocytes. Numbers of astrocytes in the periventricular white matter were correlated with increased FA and reduced ODI signal intensities. Astrocyte coherence was associated with increased FA. Moderate astrogliosis, but not loss of total oligodendrocytes, after progressive fetal inflammation can be detected with high-field diffusion tensor MRI.
Collapse
Affiliation(s)
- Robert Galinsky
- Department of Physiology, University of Auckland, Auckland 1023, New Zealand; (R.G.); (N.M.); (J.M.D.); (S.K.D.); (K.Y.); (C.A.L.); (G.W.); (J.O.D.); (V.J.K.); (L.B.)
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Victoria 3168, Australia; (F.N.); (V.A.Z.); (S.B.K.)
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, Victoria 3800, Australia
| | - Yohan van de Looij
- Division of Child Development & Growth, Department of Pediatrics, Gynaecology & Obstetrics, School of Medicine, University of Geneva, 1015 Geneva, Switzerland; (Y.v.d.L.); (S.V.S.)
| | - Natasha Mitchell
- Department of Physiology, University of Auckland, Auckland 1023, New Zealand; (R.G.); (N.M.); (J.M.D.); (S.K.D.); (K.Y.); (C.A.L.); (G.W.); (J.O.D.); (V.J.K.); (L.B.)
| | - Justin M. Dean
- Department of Physiology, University of Auckland, Auckland 1023, New Zealand; (R.G.); (N.M.); (J.M.D.); (S.K.D.); (K.Y.); (C.A.L.); (G.W.); (J.O.D.); (V.J.K.); (L.B.)
| | - Simerdeep K. Dhillon
- Department of Physiology, University of Auckland, Auckland 1023, New Zealand; (R.G.); (N.M.); (J.M.D.); (S.K.D.); (K.Y.); (C.A.L.); (G.W.); (J.O.D.); (V.J.K.); (L.B.)
| | - Kyohei Yamaguchi
- Department of Physiology, University of Auckland, Auckland 1023, New Zealand; (R.G.); (N.M.); (J.M.D.); (S.K.D.); (K.Y.); (C.A.L.); (G.W.); (J.O.D.); (V.J.K.); (L.B.)
| | - Christopher A. Lear
- Department of Physiology, University of Auckland, Auckland 1023, New Zealand; (R.G.); (N.M.); (J.M.D.); (S.K.D.); (K.Y.); (C.A.L.); (G.W.); (J.O.D.); (V.J.K.); (L.B.)
| | - Guido Wassink
- Department of Physiology, University of Auckland, Auckland 1023, New Zealand; (R.G.); (N.M.); (J.M.D.); (S.K.D.); (K.Y.); (C.A.L.); (G.W.); (J.O.D.); (V.J.K.); (L.B.)
| | - Joanne O. Davidson
- Department of Physiology, University of Auckland, Auckland 1023, New Zealand; (R.G.); (N.M.); (J.M.D.); (S.K.D.); (K.Y.); (C.A.L.); (G.W.); (J.O.D.); (V.J.K.); (L.B.)
| | - Fraser Nott
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Victoria 3168, Australia; (F.N.); (V.A.Z.); (S.B.K.)
| | - Valerie A. Zahra
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Victoria 3168, Australia; (F.N.); (V.A.Z.); (S.B.K.)
| | - Sharmony B. Kelly
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Victoria 3168, Australia; (F.N.); (V.A.Z.); (S.B.K.)
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, Victoria 3800, Australia
| | - Victoria J. King
- Department of Physiology, University of Auckland, Auckland 1023, New Zealand; (R.G.); (N.M.); (J.M.D.); (S.K.D.); (K.Y.); (C.A.L.); (G.W.); (J.O.D.); (V.J.K.); (L.B.)
| | - Stéphane V. Sizonenko
- Division of Child Development & Growth, Department of Pediatrics, Gynaecology & Obstetrics, School of Medicine, University of Geneva, 1015 Geneva, Switzerland; (Y.v.d.L.); (S.V.S.)
| | - Laura Bennet
- Department of Physiology, University of Auckland, Auckland 1023, New Zealand; (R.G.); (N.M.); (J.M.D.); (S.K.D.); (K.Y.); (C.A.L.); (G.W.); (J.O.D.); (V.J.K.); (L.B.)
| | - Alistair J. Gunn
- Department of Physiology, University of Auckland, Auckland 1023, New Zealand; (R.G.); (N.M.); (J.M.D.); (S.K.D.); (K.Y.); (C.A.L.); (G.W.); (J.O.D.); (V.J.K.); (L.B.)
- Correspondence:
| |
Collapse
|
105
|
Rollins CK, Ortinau CM, Stopp C, Friedman KG, Tworetzky W, Gagoski B, Velasco-Annis C, Afacan O, Vasung L, Beaute JI, Rofeberg V, Estroff JA, Grant PE, Soul JS, Yang E, Wypij D, Gholipour A, Warfield SK, Newburger JW. Regional Brain Growth Trajectories in Fetuses with Congenital Heart Disease. Ann Neurol 2020; 89:143-157. [PMID: 33084086 DOI: 10.1002/ana.25940] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 10/14/2020] [Accepted: 10/16/2020] [Indexed: 01/02/2023]
Abstract
OBJECTIVE Congenital heart disease (CHD) is associated with abnormal brain development in utero. We applied innovative fetal magnetic resonance imaging (MRI) techniques to determine whether reduced fetal cerebral substrate delivery impacts the brain globally, or in a region-specific pattern. Our novel design included two control groups, one with and the other without a family history of CHD, to explore the contribution of shared genes and/or fetal environment to brain development. METHODS From 2014 to 2018, we enrolled 179 pregnant women into 4 groups: "HLHS/TGA" fetuses with hypoplastic left heart syndrome (HLHS) or transposition of the great arteries (TGA), diagnoses with lowest fetal cerebral substrate delivery; "CHD-other," with other CHD diagnoses; "CHD-related," healthy with a CHD family history; and "optimal control," healthy without a family history. Two MRIs were obtained between 18 and 40 weeks gestation. Random effect regression models assessed group differences in brain volumes and relationships to hemodynamic variables. RESULTS HLHS/TGA (n = 24), CHD-other (50), and CHD-related (34) groups each had generally smaller brain volumes than the optimal controls (71). Compared with CHD-related, the HLHS/TGA group had smaller subplate (-13.3% [standard error = 4.3%], p < 0.01) and intermediate (-13.7% [4.3%], p < 0.01) zones, with a similar trend in ventricular zone (-7.1% [1.9%], p = 0.07). These volumetric reductions were associated with lower cerebral substrate delivery. INTERPRETATION Fetuses with CHD, especially those with lowest cerebral substrate delivery, show a region-specific pattern of small brain volumes and impaired brain growth before 32 weeks gestation. The brains of fetuses with CHD were more similar to those of CHD-related than optimal controls, suggesting genetic or environmental factors also contribute. ANN NEUROL 2021;89:143-157.
Collapse
Affiliation(s)
- Caitlin K Rollins
- Department of Neurology, Boston Children's Hospital, Boston, MA, USA.,Departments of Neurology, Harvard Medical School, Boston, MA, USA
| | - Cynthia M Ortinau
- Department of Pediatrics, Washington University in St. Louis, St. Louis, MO, USA
| | - Christian Stopp
- Department of Cardiology, Boston Children's Hospital, Boston, MA, USA
| | - Kevin G Friedman
- Department of Cardiology, Boston Children's Hospital, Boston, MA, USA.,Maternal Fetal Care Center, Boston Children's Hospital, Boston, MA, USA.,Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Wayne Tworetzky
- Department of Cardiology, Boston Children's Hospital, Boston, MA, USA.,Maternal Fetal Care Center, Boston Children's Hospital, Boston, MA, USA.,Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Borjan Gagoski
- Department of Radiology, Boston Children's Hospital, Boston, MA, USA.,Department of Radiology, Harvard Medical School, Boston, MA, USA
| | | | - Onur Afacan
- Department of Radiology, Boston Children's Hospital, Boston, MA, USA.,Department of Radiology, Harvard Medical School, Boston, MA, USA
| | - Lana Vasung
- Department of Radiology, Boston Children's Hospital, Boston, MA, USA.,Department of Radiology, Harvard Medical School, Boston, MA, USA
| | - Jeanette I Beaute
- Department of Neurology, Boston Children's Hospital, Boston, MA, USA
| | - Valerie Rofeberg
- Department of Cardiology, Boston Children's Hospital, Boston, MA, USA
| | - Judy A Estroff
- Department of Pediatrics, Washington University in St. Louis, St. Louis, MO, USA.,Maternal Fetal Care Center, Boston Children's Hospital, Boston, MA, USA.,Department of Radiology, Boston Children's Hospital, Boston, MA, USA.,Department of Radiology, Harvard Medical School, Boston, MA, USA
| | - P Ellen Grant
- Department of Radiology, Boston Children's Hospital, Boston, MA, USA.,Department of Radiology, Harvard Medical School, Boston, MA, USA
| | - Janet S Soul
- Department of Neurology, Boston Children's Hospital, Boston, MA, USA.,Departments of Neurology, Harvard Medical School, Boston, MA, USA.,Maternal Fetal Care Center, Boston Children's Hospital, Boston, MA, USA
| | - Edward Yang
- Department of Radiology, Boston Children's Hospital, Boston, MA, USA.,Department of Radiology, Harvard Medical School, Boston, MA, USA
| | - David Wypij
- Department of Cardiology, Boston Children's Hospital, Boston, MA, USA.,Department of Pediatrics, Harvard Medical School, Boston, MA, USA.,Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Ali Gholipour
- Department of Radiology, Boston Children's Hospital, Boston, MA, USA.,Department of Radiology, Harvard Medical School, Boston, MA, USA
| | - Simon K Warfield
- Department of Radiology, Boston Children's Hospital, Boston, MA, USA.,Department of Radiology, Harvard Medical School, Boston, MA, USA
| | - Jane W Newburger
- Department of Cardiology, Boston Children's Hospital, Boston, MA, USA.,Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
106
|
Panta A, Montgomery K, Nicolas M, Mani KK, Sampath D, Sohrabji F. Mir363-3p Treatment Attenuates Long-Term Cognitive Deficits Precipitated by an Ischemic Stroke in Middle-Aged Female Rats. Front Aging Neurosci 2020; 12:586362. [PMID: 33132904 PMCID: PMC7550720 DOI: 10.3389/fnagi.2020.586362] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 09/02/2020] [Indexed: 01/29/2023] Open
Abstract
Cognitive impairment and memory loss are commonly seen after stroke and a third of patients will develop signs of dementia a year after stroke. Despite a large number of studies on the beneficial effects of neuroprotectants, few studies have examined the effects of these compounds/interventions on long-term cognitive impairment. Our previous work showed that the microRNA mir363-3p reduced infarct volume and sensory-motor impairment in the acute stage of stroke in middle-aged females but not males. Thus, the present study determined the impact of mir363-3p treatment on stroke-induced cognitive impairment in middle-aged females. Sprague–Dawley female rats (12 months of age) were subjected to middle cerebral artery occlusion (MCAo; or sham surgery) and injected (iv) with mir363-3p mimic (MCAo + mir363-3p) or scrambled oligos (MCAo + scrambled) 4 h later. Sensory-motor performance was assessed in the acute phase (2–5 days after stroke), while all other behaviors were tested 6 months after MCAo (18 months of age). Cognitive function was assessed by the novel object recognition test (declarative memory) and the Barnes maze (spatial memory). The MCAo + scrambled group showed reduced preference for a novel object after the stroke and poor learning in the spatial memory task. In contrast, mir363-3p treated animals were similar to either their baseline performance or to the sham group. Histological analysis showed significant deterioration of specific white matter tracts due to stroke, which was attenuated in mir363-3p treated animals. The present data builds on our previous finding to show that a neuroprotectant can abrogate the long-term effects of stroke.
Collapse
Affiliation(s)
- Aditya Panta
- Women's Health in Neuroscience Program, Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center College of Medicine, Texas A&M University, Bryan, TX, United States
| | - Karienn Montgomery
- Women's Health in Neuroscience Program, Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center College of Medicine, Texas A&M University, Bryan, TX, United States
| | - Marissa Nicolas
- Women's Health in Neuroscience Program, Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center College of Medicine, Texas A&M University, Bryan, TX, United States
| | - Kathiresh K Mani
- Women's Health in Neuroscience Program, Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center College of Medicine, Texas A&M University, Bryan, TX, United States
| | - Dayalan Sampath
- Women's Health in Neuroscience Program, Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center College of Medicine, Texas A&M University, Bryan, TX, United States
| | - Farida Sohrabji
- Women's Health in Neuroscience Program, Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center College of Medicine, Texas A&M University, Bryan, TX, United States
| |
Collapse
|
107
|
Schumacher M, Liere P, Ghoumari A. Progesterone and fetal-neonatal neuroprotection. Best Pract Res Clin Obstet Gynaecol 2020; 69:50-61. [PMID: 33039311 DOI: 10.1016/j.bpobgyn.2020.09.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 09/01/2020] [Indexed: 02/05/2023]
Abstract
The role of progesterone goes beyond the maintenance of pregnancy. The hormone, indeed, protects the developing fetal brain and influences its maturation. Metabolomes analyzed by mass spectrometric methods have revealed the great diversity of steroids in maternal plasma and fetal fluids, but their developmental significance remains to be investigated. Progesterone and its metabolites reach highest levels during the third trimester, when the brain growth spurt occurs: its volume triples, synaptogenesis is particularly active, and axons start to be myelinated. This developmental stage coincides with a period of great vulnerability. Studies in sheep have shown that progesterone and its metabolite allopregnanolone protect the vulnerable fetal brain. Work in rats and mice have demonstrated that progesterone plays an important role in myelin formation. These experimental studies are discussed in relation to preterm birth. Influences of progesterone on very early stages of neural development at the beginning of pregnancy are yet to be explored.
Collapse
Affiliation(s)
- Michael Schumacher
- U1195 "Diseases and Hormones of the Nervous System", Inserm and University Paris-Saclay, 80, Rue Du Général Leclerc, 94276, Kremlin-Bicêtre, France.
| | - Philippe Liere
- U1195 "Diseases and Hormones of the Nervous System", Inserm and University Paris-Saclay, 80, Rue Du Général Leclerc, 94276, Kremlin-Bicêtre, France
| | - Abdelmoumen Ghoumari
- U1195 "Diseases and Hormones of the Nervous System", Inserm and University Paris-Saclay, 80, Rue Du Général Leclerc, 94276, Kremlin-Bicêtre, France
| |
Collapse
|
108
|
Hamdy N, Eide S, Sun HS, Feng ZP. Animal models for neonatal brain injury induced by hypoxic ischemic conditions in rodents. Exp Neurol 2020; 334:113457. [PMID: 32889009 DOI: 10.1016/j.expneurol.2020.113457] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 08/28/2020] [Accepted: 08/30/2020] [Indexed: 02/06/2023]
Abstract
Neonatal hypoxia-ischemia and resulting encephalopathies are of significant concern. Intrapartum asphyxia is a leading cause of neonatal death globally. Among surviving infants, there remains a high incidence of hypoxic-ischemic encephalopathy due to neonatal hypoxic-ischemic brain injury, manifesting as mild conditions including attention deficit hyperactivity disorder, and debilitating disorders such as cerebral palsy. Various animal models of neonatal hypoxic brain injury have been implemented to explore cellular and molecular mechanisms, assess the potential of novel therapeutic strategies, and characterize the functional and behavioural correlates of injury. Each of the animal models has individual advantages and limitations. The present review looks at several widely-used and alternative rodent models of neonatal hypoxia and hypoxia-ischemia; it highlights their strengths and limitations, and their potential for continued and improved use.
Collapse
Affiliation(s)
- Nancy Hamdy
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Sarah Eide
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Hong-Shuo Sun
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada; Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada.
| | - Zhong-Ping Feng
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada.
| |
Collapse
|
109
|
Baker EK, Jacobs SE, Lim R, Wallace EM, Davis PG. Cell therapy for the preterm infant: promise and practicalities. Arch Dis Child Fetal Neonatal Ed 2020; 105:563-568. [PMID: 32253200 DOI: 10.1136/archdischild-2019-317896] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 03/24/2020] [Accepted: 03/25/2020] [Indexed: 02/07/2023]
Abstract
Recent decades have seen the rapid progress of neonatal intensive care, and the survival rates of the most preterm infants are improving. This improvement is associated with changing patterns of morbidity and new phenotypes of bronchopulmonary dysplasia and preterm brain injury are recognised. Inflammation and immaturity are known contributors to their pathogenesis. However, a new phenomenon, the exhaustion of progenitor cells is emerging as an important factor. Current therapeutic approaches do not adequately address these new mechanisms of injury. Cell therapy, that is the use of stem and stem-like cells, with its potential to both repair and prevent injury, offers a new approach to these challenging conditions. This review will examine the rationale for cell therapy in the extremely preterm infant, the preclinical and early clinical evidence to support its use in bronchopulmonary dysplasia and preterm brain injury. Finally, it will address the challenges in translating cell therapy from the laboratory to early clinical trials.
Collapse
Affiliation(s)
- Elizabeth K Baker
- Newborn Research Centre, Royal Women's Hospital, Parkville, Victoria, Australia .,Department of Obstetrics and Gynaecology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Susan E Jacobs
- Newborn Research Centre, Royal Women's Hospital, Parkville, Victoria, Australia.,Department of Obstetrics and Gynaecology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Rebecca Lim
- Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia.,The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Euan M Wallace
- Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia.,The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Peter G Davis
- Newborn Research Centre, Royal Women's Hospital, Parkville, Victoria, Australia.,Department of Obstetrics and Gynaecology, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
110
|
Fischer I, Barak B. Molecular and Therapeutic Aspects of Hyperbaric Oxygen Therapy in Neurological Conditions. Biomolecules 2020; 10:E1247. [PMID: 32867291 PMCID: PMC7564723 DOI: 10.3390/biom10091247] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 08/19/2020] [Accepted: 08/20/2020] [Indexed: 02/07/2023] Open
Abstract
In hyperbaric oxygen therapy (HBOT), the subject is placed in a chamber containing 100% oxygen gas at a pressure of more than one atmosphere absolute. This treatment is used to hasten tissue recovery and improve its physiological aspects, by providing an increased supply of oxygen to the damaged tissue. In this review, we discuss the consequences of hypoxia, as well as the molecular and physiological processes that occur in subjects exposed to HBOT. We discuss the efficacy of HBOT in treating neurological conditions and neurodevelopmental disorders in both humans and animal models. We summarize by discussing the challenges in this field, and explore future directions that will allow the scientific community to better understand the molecular aspects and applications of HBOT for a wide variety of neurological conditions.
Collapse
Affiliation(s)
- Inbar Fischer
- The Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel;
| | - Boaz Barak
- The Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel;
- The School of Psychological Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| |
Collapse
|
111
|
Hedderich DM, Avram M, Menegaux A, Nuttall R, Zimmermann J, Schneider SC, Schmitz-Koep B, Daamen M, Scheef L, Boecker H, Zimmer C, Baumann N, Bartmann P, Wolke D, Bäuml JG, Sorg C. Hippocampal subfield volumes are nonspecifically reduced in premature-born adults. Hum Brain Mapp 2020; 41:5215-5227. [PMID: 32845045 PMCID: PMC7670635 DOI: 10.1002/hbm.25187] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 07/07/2020] [Accepted: 08/11/2020] [Indexed: 01/06/2023] Open
Abstract
Reduced global hippocampus volumes have been demonstrated in premature‐born individuals, from newborns to adults; however, it is unknown whether hippocampus subfield (HCSF) volumes are differentially affected by premature birth and how relevant they are for cognitive performance. To address these questions, we investigated magnetic resonance imaging (MRI)‐derived HCSF volumes in very premature‐born adults, and related them with general cognitive performance in adulthood. We assessed 103 very premature‐born (gestational age [GA] <32 weeks and/or birth weight <1,500 g) and 109 term‐born individuals with cognitive testing and structural MRI at 26 years of age. HCSFs were automatically segmented based on three‐dimensional T1‐ and T2‐weighted sequences and studied both individually and grouped into three functional units, namely hippocampus proper (HP), subicular complex (SC), and dentate gyrus (DG). Cognitive performance was measured using the Wechsler‐Adult‐Intelligence‐Scale (full‐scale intelligence quotient [FS‐IQ]) at 26 years. We observed bilateral volume reductions for almost all HCSF volumes in premature‐born adults and associations with GA and neonatal treatment intensity but not birth weight. Left‐sided HP, SC, and DG volumes were associated with adult FS‐IQ. Furthermore, left DG volume was a mediator of the association between GA and adult FS‐IQ in premature‐born individuals. Results demonstrate nonspecifically reduced HCSF volumes in premature‐born adults; but specific associations with cognitive outcome highlight the importance of the left DG. Data suggest that specific interventions toward hippocampus function might be promising to lower adverse cognitive effects of prematurity.
Collapse
Affiliation(s)
- Dennis M Hedderich
- Department of Neuroradiology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany.,Technical University of Munich-NIC Neuroimaging Center, Munich, Germany
| | - Mihai Avram
- Department of Neuroradiology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany.,Technical University of Munich-NIC Neuroimaging Center, Munich, Germany
| | - Aurore Menegaux
- Department of Neuroradiology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany.,Technical University of Munich-NIC Neuroimaging Center, Munich, Germany
| | - Rachel Nuttall
- Department of Neuroradiology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany.,Technical University of Munich-NIC Neuroimaging Center, Munich, Germany
| | - Juliana Zimmermann
- Department of Neuroradiology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany.,Technical University of Munich-NIC Neuroimaging Center, Munich, Germany
| | - Sebastian C Schneider
- Department of Neuroradiology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany.,Technical University of Munich-NIC Neuroimaging Center, Munich, Germany
| | - Benita Schmitz-Koep
- Department of Neuroradiology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany.,Technical University of Munich-NIC Neuroimaging Center, Munich, Germany
| | - Marcel Daamen
- Functional Neuroimaging Group, Department of Radiology, University Hospital Bonn, Bonn, Germany.,Department of Neonatology, University Hospital Bonn, Bonn, Germany
| | - Lukas Scheef
- Functional Neuroimaging Group, Department of Radiology, University Hospital Bonn, Bonn, Germany
| | - Henning Boecker
- Functional Neuroimaging Group, Department of Radiology, University Hospital Bonn, Bonn, Germany
| | - Claus Zimmer
- Department of Neuroradiology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany.,Technical University of Munich-NIC Neuroimaging Center, Munich, Germany
| | - Nicole Baumann
- Department of Psychology, University of Warwick, Coventry, UK.,Warwick Medical School, University of Warwick, Coventry, UK
| | - Peter Bartmann
- Department of Neonatology, University Hospital Bonn, Bonn, Germany
| | - Dieter Wolke
- Department of Psychology, University of Warwick, Coventry, UK.,Warwick Medical School, University of Warwick, Coventry, UK
| | - Josef G Bäuml
- Department of Neuroradiology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany.,Technical University of Munich-NIC Neuroimaging Center, Munich, Germany
| | - Christian Sorg
- Department of Neuroradiology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany.,Technical University of Munich-NIC Neuroimaging Center, Munich, Germany.,Department of Psychiatry, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| |
Collapse
|
112
|
Sunny DE, Hammer E, Strempel S, Joseph C, Manchanda H, Ittermann T, Hübner S, Weiss FU, Völker U, Heckmann M. Nup133 and ERα mediate the differential effects of hyperoxia-induced damage in male and female OPCs. Mol Cell Pediatr 2020; 7:10. [PMID: 32844334 PMCID: PMC7447710 DOI: 10.1186/s40348-020-00102-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 08/02/2020] [Indexed: 02/07/2023] Open
Abstract
Background Hyperoxia is a well-known cause of cerebral white matter injury in preterm infants with male sex being an independent and critical risk factor for poor neurodevelopmental outcome. Sex is therefore being widely considered as one of the major decisive factors for prognosis and treatment of these infants. But unfortunately, we still lack a clear view of the molecular mechanisms that lead to such a profound difference. Hence, using mouse-derived primary oligodendrocyte progenitor cells (OPCs), we investigated the molecular factors and underlying mechanisms behind the differential response of male and female cells towards oxidative stress. Results We demonstrate that oxidative stress severely affects cellular functions related to energy metabolism, stress response, and maturation in the male-derived OPCs, whereas the female cells remain largely unaffected. CNPase protein level was found to decline following hyperoxia in male but not in female cells. This impairment of maturation was accompanied by the downregulation of nucleoporin and nuclear lamina proteins in the male cells. We identify Nup133 as a novel target protein affected by hyperoxia, whose inverse regulation may mediate this differential response in the male and female cells. Nup133 protein level declined following hyperoxia in male but not in female cells. We show that nuclear respiratory factor 1 (Nrf1) is a direct downstream target of Nup133 and that Nrf1 mRNA declines following hyperoxia in male but not in female cells. The female cells may be rendered resistant due to synergistic protection via the estrogen receptor alpha (ERα) which was upregulated following hyperoxia in female but not in male cells. Both Nup133 and ERα regulate mitochondrial function and oxidative stress response by transcriptional regulation of Nrf1. Conclusions These findings from a basic cell culture model establish prominent sex-based differences and suggest a novel mechanism involved in the differential response of OPCs towards oxidative stress. It conveys a strong message supporting the need to study how complex cellular processes are regulated differently in male and female brains during development and for a better understanding of how the brain copes up with different forms of stress after preterm birth.
Collapse
Affiliation(s)
- Donna Elizabeth Sunny
- Department of Neonatology and Pediatric Intensive Care, University of Medicine Greifswald, Ferdinand-Sauerbruchstrasse, 17475, Greifswald, Germany.
| | - Elke Hammer
- Department of Functional Genomics, University of Medicine Greifswald, Greifswald, Germany
| | | | - Christy Joseph
- Department of Pharmacology, Center of Drug Absorption and Transport (C_DAT), University of Medicine Greifswald, Greifswald, Germany
| | - Himanshu Manchanda
- Department of Bioinformatics, University of Medicine Greifswald, Greifswald, Germany
| | - Till Ittermann
- Institute for Community Medicine, University of Medicine Greifswald, Greifswald, Germany
| | - Stephanie Hübner
- Department of Neonatology and Pediatric Intensive Care, University of Medicine Greifswald, Ferdinand-Sauerbruchstrasse, 17475, Greifswald, Germany
| | - Frank Ulrich Weiss
- Department of Internal Medicine A, University of Medicine Greifswald, Greifswald, Germany
| | - Uwe Völker
- Department of Functional Genomics, University of Medicine Greifswald, Greifswald, Germany
| | - Matthias Heckmann
- Department of Neonatology and Pediatric Intensive Care, University of Medicine Greifswald, Ferdinand-Sauerbruchstrasse, 17475, Greifswald, Germany
| |
Collapse
|
113
|
Schmitz-Koep B, Bäuml JG, Menegaux A, Nuttall R, Zimmermann J, Schneider SC, Daamen M, Scheef L, Boecker H, Zimmer C, Gaser C, Wolke D, Bartmann P, Sorg C, Hedderich DM. Decreased cortical thickness mediates the relationship between premature birth and cognitive performance in adulthood. Hum Brain Mapp 2020; 41:4952-4963. [PMID: 32820839 PMCID: PMC7643384 DOI: 10.1002/hbm.25172] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 06/25/2020] [Accepted: 08/04/2020] [Indexed: 01/05/2023] Open
Abstract
Cortical thickness (CTh) reflects cortical properties such as dendritic complexity and synaptic density, which are not only vulnerable to developmental disturbances caused by premature birth but also highly relevant for cognitive performance. We tested the hypotheses whether CTh in young adults is altered after premature birth and whether these aberrations are relevant for general cognitive abilities. We investigated CTh based on brain structural magnetic resonance imaging and surface-based morphometry in a large and prospectively collected cohort of 101 very premature-born adults (<32 weeks of gestation and/or birth weight [BW] below 1,500 g) and 111 full-term controls at 26 years of age. Cognitive performance was assessed by full-scale intelligence quotient (IQ) using the Wechsler Adult Intelligence Scale. CTh was reduced in frontal, parietal, and temporal associative cortices predominantly in the left hemisphere in premature-born adults compared to controls. We found a significant positive association of CTh with both gestational age and BW, particularly in the left hemisphere, and a significant negative association between CTh and intensity of neonatal treatment within limited regions bilaterally. Full-scale IQ and CTh in the left hemisphere were positively correlated. Furthermore, CTh in the left hemisphere acted as a mediator on the association between premature birth and full-scale IQ. Results provide evidence that premature born adults have widespread reduced CTh that is relevant for their general cognitive performance. Data suggest lasting reductions in cortical microstructure subserving CTh after premature birth.
Collapse
Affiliation(s)
- Benita Schmitz-Koep
- Department of Neuroradiology, School of Medicine, Technical University of Munich, Munich, Germany.,TUM-NIC Neuroimaging Center, School of Medicine, Technical University of Munich, Munich, Germany
| | - Josef G Bäuml
- Department of Neuroradiology, School of Medicine, Technical University of Munich, Munich, Germany.,TUM-NIC Neuroimaging Center, School of Medicine, Technical University of Munich, Munich, Germany
| | - Aurore Menegaux
- Department of Neuroradiology, School of Medicine, Technical University of Munich, Munich, Germany.,TUM-NIC Neuroimaging Center, School of Medicine, Technical University of Munich, Munich, Germany
| | - Rachel Nuttall
- Department of Neuroradiology, School of Medicine, Technical University of Munich, Munich, Germany.,TUM-NIC Neuroimaging Center, School of Medicine, Technical University of Munich, Munich, Germany
| | - Juliana Zimmermann
- Department of Neuroradiology, School of Medicine, Technical University of Munich, Munich, Germany.,TUM-NIC Neuroimaging Center, School of Medicine, Technical University of Munich, Munich, Germany
| | - Sebastian C Schneider
- Department of Neuroradiology, School of Medicine, Technical University of Munich, Munich, Germany.,TUM-NIC Neuroimaging Center, School of Medicine, Technical University of Munich, Munich, Germany
| | - Marcel Daamen
- Functional Neuroimaging Group, Department of Radiology, University Hospital Bonn, Bonn, Germany.,Department of Neonatology, University Hospital Bonn, Bonn, Germany
| | - Lukas Scheef
- Functional Neuroimaging Group, Department of Radiology, University Hospital Bonn, Bonn, Germany
| | - Henning Boecker
- Functional Neuroimaging Group, Department of Radiology, University Hospital Bonn, Bonn, Germany
| | - Claus Zimmer
- TUM-NIC Neuroimaging Center, School of Medicine, Technical University of Munich, Munich, Germany
| | - Christian Gaser
- Department of Psychiatry, University Hospital Jena, Jena, Germany.,Department of Neurology, University Hospital Jena, Jena, Germany
| | - Dieter Wolke
- Department of Psychology, University of Warwick, Coventry, UK.,Warwick Medical School, University of Warwick, Coventry, UK
| | - Peter Bartmann
- Department of Neonatology, University Hospital Bonn, Bonn, Germany
| | - Christian Sorg
- Department of Neuroradiology, School of Medicine, Technical University of Munich, Munich, Germany.,TUM-NIC Neuroimaging Center, School of Medicine, Technical University of Munich, Munich, Germany.,Department of Psychiatry, School of Medicine, Technical University of Munich, Munich, Germany
| | - Dennis M Hedderich
- Department of Neuroradiology, School of Medicine, Technical University of Munich, Munich, Germany.,TUM-NIC Neuroimaging Center, School of Medicine, Technical University of Munich, Munich, Germany
| |
Collapse
|
114
|
Carlström KE, Zhu K, Ewing E, Krabbendam IE, Harris RA, Falcão AM, Jagodic M, Castelo-Branco G, Piehl F. Gsta4 controls apoptosis of differentiating adult oligodendrocytes during homeostasis and remyelination via the mitochondria-associated Fas-Casp8-Bid-axis. Nat Commun 2020; 11:4071. [PMID: 32792491 PMCID: PMC7426940 DOI: 10.1038/s41467-020-17871-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 07/23/2020] [Indexed: 01/20/2023] Open
Abstract
Arrest of oligodendrocyte (OL) differentiation and remyelination following myelin damage in multiple sclerosis (MS) is associated with neurodegeneration and clinical worsening. We show that Glutathione S-transferase 4α (Gsta4) is highly expressed during adult OL differentiation and that Gsta4 loss impairs differentiation into myelinating OLs in vitro. In addition, we identify Gsta4 as a target of both dimethyl fumarate, an existing MS therapy, and clemastine fumarate, a candidate remyelinating agent in MS. Overexpression of Gsta4 reduces expression of Fas and activity of the mitochondria-associated Casp8-Bid-axis in adult oligodendrocyte precursor cells, leading to improved OL survival during differentiation. The Gsta4 effect on apoptosis during adult OL differentiation was corroborated in vivo in both lysolecithin-induced demyelination and experimental autoimmune encephalomyelitis models, where Casp8 activity was reduced in Gsta4-overexpressing OLs. Our results identify Gsta4 as an intrinsic regulator of OL differentiation, survival and remyelination, as well as a potential target for future reparative MS therapies.
Collapse
Affiliation(s)
- Karl E Carlström
- Department of Clinical Neurosciences, Karolinska Institutet, Center for Molecular Medicine, Karolinska Hospital at Solna, 17177, Stockholm, Sweden.
| | - Keying Zhu
- Department of Clinical Neurosciences, Karolinska Institutet, Center for Molecular Medicine, Karolinska Hospital at Solna, 17177, Stockholm, Sweden
| | - Ewoud Ewing
- Department of Clinical Neurosciences, Karolinska Institutet, Center for Molecular Medicine, Karolinska Hospital at Solna, 17177, Stockholm, Sweden
| | - Inge E Krabbendam
- Department of Clinical Neurosciences, Karolinska Institutet, Center for Molecular Medicine, Karolinska Hospital at Solna, 17177, Stockholm, Sweden
| | - Robert A Harris
- Department of Clinical Neurosciences, Karolinska Institutet, Center for Molecular Medicine, Karolinska Hospital at Solna, 17177, Stockholm, Sweden
| | - Ana Mendanha Falcão
- Laboratory of Molecular Neurobiology, Department Medical Biochemistry and Biophysics, Biomedicum, Karolinska Institutet, 17177, Stockholm, Sweden
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Minho, Portugal
| | - Maja Jagodic
- Department of Clinical Neurosciences, Karolinska Institutet, Center for Molecular Medicine, Karolinska Hospital at Solna, 17177, Stockholm, Sweden
| | - Gonçalo Castelo-Branco
- Laboratory of Molecular Neurobiology, Department Medical Biochemistry and Biophysics, Biomedicum, Karolinska Institutet, 17177, Stockholm, Sweden
- Ming Wai Lau Centre for Reparative Medicine, Stockholm node, Karolinska Institutet, Stockholm, Sweden
| | - Fredrik Piehl
- Department of Clinical Neurosciences, Karolinska Institutet, Center for Molecular Medicine, Karolinska Hospital at Solna, 17177, Stockholm, Sweden
| |
Collapse
|
115
|
Ophelders DR, Gussenhoven R, Klein L, Jellema RK, Westerlaken RJ, Hütten MC, Vermeulen J, Wassink G, Gunn AJ, Wolfs TG. Preterm Brain Injury, Antenatal Triggers, and Therapeutics: Timing Is Key. Cells 2020; 9:E1871. [PMID: 32785181 PMCID: PMC7464163 DOI: 10.3390/cells9081871] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 07/31/2020] [Accepted: 08/05/2020] [Indexed: 02/08/2023] Open
Abstract
With a worldwide incidence of 15 million cases, preterm birth is a major contributor to neonatal mortality and morbidity, and concomitant social and economic burden Preterm infants are predisposed to life-long neurological disorders due to the immaturity of the brain. The risks are inversely proportional to maturity at birth. In the majority of extremely preterm infants (<28 weeks' gestation), perinatal brain injury is associated with exposure to multiple inflammatory perinatal triggers that include antenatal infection (i.e., chorioamnionitis), hypoxia-ischemia, and various postnatal injurious triggers (i.e., oxidative stress, sepsis, mechanical ventilation, hemodynamic instability). These perinatal insults cause a self-perpetuating cascade of peripheral and cerebral inflammation that plays a critical role in the etiology of diffuse white and grey matter injuries that underlies a spectrum of connectivity deficits in survivors from extremely preterm birth. This review focuses on chorioamnionitis and hypoxia-ischemia, which are two important antenatal risk factors for preterm brain injury, and highlights the latest insights on its pathophysiology, potential treatment, and future perspectives to narrow the translational gap between preclinical research and clinical applications.
Collapse
Affiliation(s)
- Daan R.M.G. Ophelders
- Department of Pediatrics, Maastricht University Medical Center, 6202 AZ Maastricht, The Netherlands; (D.R.M.G.O.); (R.G.); (L.K.); (R.K.J.); (R.J.J.W.); (M.C.H.)
- School for Oncology and Developmental Biology (GROW), Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Ruth Gussenhoven
- Department of Pediatrics, Maastricht University Medical Center, 6202 AZ Maastricht, The Netherlands; (D.R.M.G.O.); (R.G.); (L.K.); (R.K.J.); (R.J.J.W.); (M.C.H.)
| | - Luise Klein
- Department of Pediatrics, Maastricht University Medical Center, 6202 AZ Maastricht, The Netherlands; (D.R.M.G.O.); (R.G.); (L.K.); (R.K.J.); (R.J.J.W.); (M.C.H.)
- School for Mental Health and Neuroscience (MHeNS), Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Reint K. Jellema
- Department of Pediatrics, Maastricht University Medical Center, 6202 AZ Maastricht, The Netherlands; (D.R.M.G.O.); (R.G.); (L.K.); (R.K.J.); (R.J.J.W.); (M.C.H.)
| | - Rob J.J. Westerlaken
- Department of Pediatrics, Maastricht University Medical Center, 6202 AZ Maastricht, The Netherlands; (D.R.M.G.O.); (R.G.); (L.K.); (R.K.J.); (R.J.J.W.); (M.C.H.)
- School for Oncology and Developmental Biology (GROW), Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Matthias C. Hütten
- Department of Pediatrics, Maastricht University Medical Center, 6202 AZ Maastricht, The Netherlands; (D.R.M.G.O.); (R.G.); (L.K.); (R.K.J.); (R.J.J.W.); (M.C.H.)
- School for Oncology and Developmental Biology (GROW), Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Jeroen Vermeulen
- Department of Pediatric Neurology, Maastricht University Medical Center, 6202 AZ Maastricht, The Netherlands;
| | - Guido Wassink
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Private bag 92019, Auckland 1023, New Zealand; (G.W.); (A.J.G.)
| | - Alistair J. Gunn
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Private bag 92019, Auckland 1023, New Zealand; (G.W.); (A.J.G.)
| | - Tim G.A.M. Wolfs
- Department of Pediatrics, Maastricht University Medical Center, 6202 AZ Maastricht, The Netherlands; (D.R.M.G.O.); (R.G.); (L.K.); (R.K.J.); (R.J.J.W.); (M.C.H.)
- School for Oncology and Developmental Biology (GROW), Maastricht University, 6229 ER Maastricht, The Netherlands
| |
Collapse
|
116
|
Yap V, Perlman JM. Mechanisms of brain injury in newborn infants associated with the fetal inflammatory response syndrome. Semin Fetal Neonatal Med 2020; 25:101110. [PMID: 32303463 DOI: 10.1016/j.siny.2020.101110] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The fetal inflammatory response syndrome (FIRS) is characterized by umbilical cord inflammation and elevated fetal pro-inflammatory cytokines. Surviving neonates, especially very preterm infants, have increased rates of neonatal morbidity including neurodevelopmental impairment. The mechanism of brain injury in FIRS is complex and may involve "multiple hits." Exposure to in utero inflammation initiates a cascade of the fetal immune response, where pro-inflammatory cytokines can cause direct injury to oligodendrocytes and neurons. Activation of microglia results in further injury to vulnerable pre-myelinating oligodendrocytes and influences the integrity of the fetal and newborn's blood-brain barrier, resulting in further exposure of the brain to developmental insults. Newborns exposed to FIRS are frequently exposed to additional perinatal and postnatal insults that can result in further brain injury. Future directions should include evaluations for new therapeutic interventions aimed at reducing brain injury by dampening FIRS, inhibition of microglial activation, and regeneration of immature oligodendrocytes.
Collapse
Affiliation(s)
- Vivien Yap
- Weill Cornell Medicine - New York Presbyterian Hospital, 525 East 68th Street, Suite N-506, New York, NY, 10065, United States.
| | - Jeffrey M Perlman
- Weill Cornell Medicine - New York Presbyterian Hospital, 525 East 68th Street, Suite N-506, New York, NY, 10065, United States
| |
Collapse
|
117
|
Ortinau CM, Shimony JS. The Congenital Heart Disease Brain: Prenatal Considerations for Perioperative Neurocritical Care. Pediatr Neurol 2020; 108:23-30. [PMID: 32107137 PMCID: PMC7306416 DOI: 10.1016/j.pediatrneurol.2020.01.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 12/21/2019] [Accepted: 01/05/2020] [Indexed: 12/17/2022]
Abstract
Altered brain development has been highlighted as an important contributor to adverse neurodevelopmental outcomes in children with congenital heart disease. Abnormalities begin prenatally and include micro- and macrostructural disturbances that lead to an altered trajectory of brain growth throughout gestation. Recent progress in fetal imaging has improved understanding of the neurobiological mechanisms and risk factors for impaired fetal brain development. The impact of the prenatal environment on postnatal neurological care has also gained increased focus. This review summarizes current data on the timing and pattern of altered prenatal brain development in congenital heart disease, the potential mechanisms of these abnormalities, and the association with perioperative neurological complications.
Collapse
Affiliation(s)
- Cynthia M Ortinau
- Department of Pediatrics, Washington University in St. Louis, St. Louis, Missouri.
| | - Joshua S Shimony
- Mallinkrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri
| |
Collapse
|
118
|
Wang D, Ye X, Xie H, Liu Y, Xu Y, Wang Y, Zhou Y, Zhou N, Wang J. Profiling analysis reveals the potential contribution of long non-coding RNAs to preterm white matter injury. Life Sci 2020; 255:117815. [PMID: 32442450 DOI: 10.1016/j.lfs.2020.117815] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 05/09/2020] [Accepted: 05/15/2020] [Indexed: 12/15/2022]
Abstract
AIMS The aim of this study was to investigate the molecular mechanism underlying preterm white matter injury (WMI) via the identification and functional analysis of differentially expressed long non-coding RNAs (lncRNAs) and mRNAs. MAIN METHODS A neonatal rat model of preterm WMI was established by ligating the common carotid artery and hypoxia induction. RNA sequencing was performed to analyze gene expression profiles of brain samples. Gene Ontology (GO) and Kyoto Encyclopedia of Genes (KEGG) analyses were performed to evaluate functions of target mRNAs. A co-expression network was generated to explore regulatory mechanisms. KEY FINDINGS In total, 210 lncRNAs and 619 mRNAs were differentially expressed between the preterm WMI group and the sham group. Based on GO and KEGG analyses, enriched pathways included the apoptotic signaling pathway, vascular endothelial growth factor (VEGF) signaling pathway, natural killer cell-mediated cytotoxicity pathway, and the autophagy pathway. SIGNIFICANCE Differentially expressed lncRNAs and mRNAs in the brain tissues of preterm WMI model were identified, and the biological processes were closely associated with the development of preterm WMI, thus being considered potential targets for future studies.
Collapse
Affiliation(s)
- Dayu Wang
- The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, China
| | - Xiaoyan Ye
- The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, China
| | - Hanying Xie
- The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, China
| | - Yiwen Liu
- The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, China
| | - Yan Xu
- The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, China
| | - Yao Wang
- The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, China
| | - Yang Zhou
- The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, China
| | - Ning Zhou
- Xuzhou Medical University, Xuzhou 221000, China
| | - Jun Wang
- The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, China.
| |
Collapse
|
119
|
Abstract
Significant advances in the field of neonatal imaging has resulted in the generation of large complex data sets of relevant information for routine daily clinical practice, and basic and translational research. The evaluation of this data is a complex task for the neonatal imager who must distinguish normal and incidental findings from clinically significant abnormalities which are often adjunctive data points applicable to clinical evaluation and treatment. This review provides an overview of the imaging manifestations of disease processes commonly encountered in the neonatal brain. Since MRI is currently the highest yield technique for the diagnosis and characterization of the normal and abnormal brain, it is therefore the focus of the majority of this review. When applicable, discussion of some of the pertinent known pathophysiology and neuropathological aspects of disease processes are reviewed.
Collapse
|
120
|
Yin L, Wang S, Zhang N, Bai X, Xie J, Wen Q, Huang L, Qian L, Jiang L. Elevation of stromal cell-derived factor 1 and C-X-C chemokine receptor type 4 in white matter damage treatment with recombinant human erythropoietin and human umbilical cord mesenchymal stem cells in a rat model of preterm birth. Int J Dev Neurosci 2020; 80:247-256. [PMID: 32108377 DOI: 10.1002/jdn.10021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 02/19/2020] [Accepted: 02/24/2020] [Indexed: 11/05/2022] Open
Abstract
OBJECTIVES To investigate the role of stromal cell-derived factor 1 (SDF-1) and C-X-C chemokine receptor type 4 (CXCR-4) in the premature brain with white matter damage (WMD) undergoing treatment with human umbilical cord mesenchymal stem cells (hUC-MSCs) and recombinant human erythropoietin (rhEPO). EXPERIMENTAL DESIGN Three-day-old Sprague-Dawley (SD) rats were randomly divided into sham operation group, hypoxia-ischemia (HI) group, rhEPO treated HI group, hUC-MSCs treated HI group, and rhEPO + hUC-MSCs treated HI group. WMD was established in all groups except the Sham group. SDF-1 and CXCR-4 levels in each group were detected at postnatal day (P) 5, P7, and P14. Pathological changes were assessed via HE staining at P14 and neuroethological tests were performed at P28. OBSERVATIONS AND CONCLUSIONS The rhEPO and hUC-MSCs intervention reduced injury area, increased body weight at P7, and improved neurobehavioral scores at P28. Furthermore, their combined use proved even more beneficial. SDF-1 levels in the rhEPO group were higher than those in the other groups and highest in the hUC-MSCs + rhEPO group (all p < .01). SDF-1 levels in the hUC-MSCs + rhEPO and rhEPO groups were increased at P5 and reached a peak at P7. CXCR-4 levels in the hUC-MSCs group were higher than those in the other groups and highest in the hUC-MSCs + rhEPO group (all p < .01). CXCR-4 levels were also increased at P5 and highest at P14. SIGNIFICANCE hUC-MSCs + rhEPO might reduce nerve cell damage and improve neurobehavioral development, in connection with increased SDF-1 and CXCR-4 expression, in premature rats with WMD due to hypoxic-ischemic injury.
Collapse
Affiliation(s)
- Liping Yin
- Department of Pediatrics, Zhongda Hospital, Southeast University, Nanjing, China.,Teaching and Research Section of Pediatrics, Medical College, Southeast University, Nanjing, China
| | - Shiyu Wang
- Department of Pediatrics, Xuanwu Hospital Medical University, Beijing, China
| | - Ning Zhang
- Department of Pediatrics, Zhongda Hospital, Southeast University, Nanjing, China.,Teaching and Research Section of Pediatrics, Medical College, Southeast University, Nanjing, China
| | - Xiang Bai
- Neonatal Intensive Care Unit, Gansu Provincial Maternity and Child-care Hospital, Lanzhou, China
| | - Jiali Xie
- Department of Pediatrics, Zhongda Hospital, Southeast University, Nanjing, China.,Teaching and Research Section of Pediatrics, Medical College, Southeast University, Nanjing, China
| | - Quan Wen
- Department of Pediatrics, Zhongda Hospital, Southeast University, Nanjing, China.,Teaching and Research Section of Pediatrics, Medical College, Southeast University, Nanjing, China
| | - Li Huang
- Department of Pediatrics, Zhongda Hospital, Southeast University, Nanjing, China.,Teaching and Research Section of Pediatrics, Medical College, Southeast University, Nanjing, China
| | - Lijuan Qian
- Department of Pediatrics, Zhongda Hospital, Southeast University, Nanjing, China.,Teaching and Research Section of Pediatrics, Medical College, Southeast University, Nanjing, China
| | - Li Jiang
- Department of Pediatrics, Zhongda Hospital, Southeast University, Nanjing, China.,Teaching and Research Section of Pediatrics, Medical College, Southeast University, Nanjing, China
| |
Collapse
|
121
|
Belov Kirdajova D, Kriska J, Tureckova J, Anderova M. Ischemia-Triggered Glutamate Excitotoxicity From the Perspective of Glial Cells. Front Cell Neurosci 2020; 14:51. [PMID: 32265656 PMCID: PMC7098326 DOI: 10.3389/fncel.2020.00051] [Citation(s) in RCA: 232] [Impact Index Per Article: 46.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 02/21/2020] [Indexed: 12/21/2022] Open
Abstract
A plethora of neurological disorders shares a final common deadly pathway known as excitotoxicity. Among these disorders, ischemic injury is a prominent cause of death and disability worldwide. Brain ischemia stems from cardiac arrest or stroke, both responsible for insufficient blood supply to the brain parenchyma. Glucose and oxygen deficiency disrupts oxidative phosphorylation, which results in energy depletion and ionic imbalance, followed by cell membrane depolarization, calcium (Ca2+) overload, and extracellular accumulation of excitatory amino acid glutamate. If tight physiological regulation fails to clear the surplus of this neurotransmitter, subsequent prolonged activation of glutamate receptors forms a vicious circle between elevated concentrations of intracellular Ca2+ ions and aberrant glutamate release, aggravating the effect of this ischemic pathway. The activation of downstream Ca2+-dependent enzymes has a catastrophic impact on nervous tissue leading to cell death, accompanied by the formation of free radicals, edema, and inflammation. After decades of “neuron-centric” approaches, recent research has also finally shed some light on the role of glial cells in neurological diseases. It is becoming more and more evident that neurons and glia depend on each other. Neuronal cells, astrocytes, microglia, NG2 glia, and oligodendrocytes all have their roles in what is known as glutamate excitotoxicity. However, who is the main contributor to the ischemic pathway, and who is the unsuspecting victim? In this review article, we summarize the so-far-revealed roles of cells in the central nervous system, with particular attention to glial cells in ischemia-induced glutamate excitotoxicity, its origins, and consequences.
Collapse
Affiliation(s)
- Denisa Belov Kirdajova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic (ASCR), Prague, Czechia.,Second Faculty of Medicine, Charles University, Prague, Czechia
| | - Jan Kriska
- Department of Cellular Neurophysiology, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic (ASCR), Prague, Czechia.,Second Faculty of Medicine, Charles University, Prague, Czechia
| | - Jana Tureckova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic (ASCR), Prague, Czechia
| | - Miroslava Anderova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic (ASCR), Prague, Czechia.,Second Faculty of Medicine, Charles University, Prague, Czechia
| |
Collapse
|
122
|
Hedderich DM, Bäuml JG, Menegaux A, Avram M, Daamen M, Zimmer C, Bartmann P, Scheef L, Boecker H, Wolke D, Gaser C, Sorg C. An analysis of MRI derived cortical complexity in premature-born adults: Regional patterns, risk factors, and potential significance. Neuroimage 2020; 208:116438. [DOI: 10.1016/j.neuroimage.2019.116438] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 11/18/2019] [Accepted: 12/03/2019] [Indexed: 01/20/2023] Open
|
123
|
Extrinsic Factors Driving Oligodendrocyte Lineage Cell Progression in CNS Development and Injury. Neurochem Res 2020; 45:630-642. [PMID: 31997102 PMCID: PMC7058689 DOI: 10.1007/s11064-020-02967-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 12/19/2019] [Accepted: 12/21/2019] [Indexed: 12/15/2022]
Abstract
Oligodendrocytes (OLs) generate myelin membranes for the rapid propagation of electrical signals along axons in the central nervous system (CNS) and provide metabolites to support axonal integrity and function. Differentiation of OLs from oligodendrocyte progenitor cells (OPCs) is orchestrated by a multitude of intrinsic and extrinsic factors in the CNS. Disruption of this process, or OL loss in the developing or adult brain, as observed in various neurological conditions including hypoxia/ischemia, stroke, and demyelination, results in axonal dystrophy, neuronal dysfunction, and severe neurological impairments. While much is known regarding the intrinsic regulatory signals required for OL lineage cell progression in development, studies from pathological conditions highlight the importance of the CNS environment and external signals in regulating OL genesis and maturation. Here, we review the recent findings in OL biology in the context of the CNS physiological and pathological conditions, focusing on extrinsic factors that facilitate OL development and regeneration.
Collapse
|
124
|
White Matter and Neuroprotection in Alzheimer's Dementia. Molecules 2020; 25:molecules25030503. [PMID: 31979414 PMCID: PMC7038211 DOI: 10.3390/molecules25030503] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 01/20/2020] [Accepted: 01/21/2020] [Indexed: 12/12/2022] Open
Abstract
Myelin is the main component of the white matter of the central nervous system (CNS), allowing the proper electrical function of the neurons by ensheathing and insulating the axons. The extensive use of magnetic resonance imaging has highlighted the white matter alterations in Alzheimer’s dementia (AD) and other neurodegenerative diseases, alterations which are early, extended, and regionally selective. Given that the white matter turnover is considerable in the adulthood, and that myelin repair is currently recognized as being the only true reparative capability of the mature CNS, oligodendrocyte precursor cells (OPCs), the cells that differentiate in oligodendrocyte, responsible for myelin formation and repair, are regarded as a potential target for neuroprotection. In this review, several aspects of the OPC biology are reviewed. The histology and functional role of OPCs in the neurovascular-neuroglial unit as described in preclinical and clinical studies on AD is discussed, such as the OPC vulnerability to hypoxia-ischemia, neuroinflammation, and amyloid deposition. Finally, the position of OPCs in drug discovery strategies for dementia is discussed.
Collapse
|
125
|
Yu Y, Luo X, Li C, Ding F, Wang M, Xie M, Yu Z, Ransom BR, Wang W. Microglial Hv1 proton channels promote white matter injuries after chronic hypoperfusion in mice. J Neurochem 2019; 152:350-367. [PMID: 31769505 DOI: 10.1111/jnc.14925] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Revised: 11/16/2019] [Accepted: 11/19/2019] [Indexed: 12/30/2022]
Abstract
Microglia are critical in damage/repair processes during ischemic white matter injury (WMI). Voltage-gated proton channel (Hv1) is expressed in microglia and contributes to nicotinamide adenine dinucleotide phosphate oxidase complex-dependent production of reactive oxygen species (ROS). Recent findings have shown that Hv1 is involved in regulating luminal pH of M1-polarized microglial phagosomes and inhibits endocytosis in microglia. We previously reported that Hv1 facilitated production of ROS and pro-inflammatory cytokines in microglia and enhanced damage to oligodendrocyte progenitor cells from oxygen and glucose deprivation. To investigate the role of Hv1 in hypoperfusion-induced WMI, we employed mice that were genetically devoid of Hv1 (Hv1-/- ), as well as a model of subcortical vascular dementia via bilateral common carotid artery stenosis. Integrity of myelin was assessed using immunofluorescent staining and transmission electron microscopy, while cognitive impairment was assessed using an eight-arm radial maze test. Hv1 deficiency was found to attenuate bilateral common carotid artery stenosis-induced disruption of white matter integrity and impairment of working memory. Immunofluorescent staining and western blotting were used to assay changes in oligodendrocytes, OPCs, and microglial polarization. Compared with that in wild-type (WT) mice, Hv1-/- mice exhibited reduced ROS generation, decreased pro-inflammatory cytokines production, and an M2-dominant rather than M1-dominant microglial polarization. Furthermore, Hv1-/- mice exhibited enhanced OPC proliferation and differentiation into oligodendrocytes. Results of mouse-derived microglia-OPC co-cultures suggested that PI3K/Akt signaling was involved in Hv1-deficiency-induced M2-type microglial polarization and concomitant OPC differentiation. These results suggest that microglial Hv1 is a promising therapeutic target for reducing ischemic WMI and cognitive impairment.
Collapse
Affiliation(s)
- Ying Yu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiang Luo
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chunyu Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fengfei Ding
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Minghuan Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Minjie Xie
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhiyuan Yu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bruce R Ransom
- Department of Neurology, University of Washington School of Medicine HMC, Seattle, WA, USA
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
126
|
Baldassarro VA, Marchesini A, Giardino L, Calzà L. Differential effects of glucose deprivation on the survival of fetal versus adult neural stem cells-derived oligodendrocyte precursor cells. Glia 2019; 68:898-917. [PMID: 31755592 DOI: 10.1002/glia.23750] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 10/28/2019] [Accepted: 10/29/2019] [Indexed: 12/27/2022]
Abstract
Impaired myelination is a key feature in neonatal hypoxia/ischemia (HI), the most common perinatal/neonatal cause of death and permanent disabilities, which is triggered by the establishment of an inflammatory and hypoxic environment during the most critical period of myelin development. This process is dependent on oligodendrocyte precursor cells (OPCs) and their capability to differentiate into mature oligodendrocytes. In this study, we investigated the vulnerability of fetal and adult OPCs derived from neural stem cells (NSCs) to inflammatory and HI insults. The resulting OPCs/astrocytes cultures were exposed to cytokines to mimic inflammation, or to oxygen-glucose deprivation (OGD) to mimic an HI condition. The differentiation of both fetal and adult OPCs is completely abolished following exposure to inflammatory cytokines, while only fetal-derived OPCs degenerate when exposed to OGD. We then investigated possible mechanisms involved in OGD-mediated toxicity: (a) T3-mediated maturation induction; (b) glutamate excitotoxicity; (c) glucose metabolism. We found that while no substantial differences were observed in T3 intracellular content regulation and glutamate-mediated toxicity, glucose deprivation lead to selective OPC cell death and impaired differentiation in fetal cultures only. These results indicate that the biological response of OPCs to inflammation and demyelination is different in fetal and adult cells, and that the glucose metabolism perturbation in fetal central nervous system (CNS) may significantly contribute to neonatal pathologies. An understanding of the underlying molecular mechanism will contribute greatly to differentiating myelination enhancing and neuroprotective therapies for neonatal and adult CNS white matter lesions.
Collapse
Affiliation(s)
- Vito Antonio Baldassarro
- Health Science and Technologies Interdepartmental Center for Industrial Research (HST-ICIR), University of Bologna, Bologna, Italy.,Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy.,Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France
| | | | - Luciana Giardino
- Health Science and Technologies Interdepartmental Center for Industrial Research (HST-ICIR), University of Bologna, Bologna, Italy.,IRET Foundation, Ozzano Emilia, Italy.,Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | - Laura Calzà
- Health Science and Technologies Interdepartmental Center for Industrial Research (HST-ICIR), University of Bologna, Bologna, Italy.,Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy.,IRET Foundation, Ozzano Emilia, Italy
| |
Collapse
|
127
|
Hirayama T, Hiraoka Y, Kitamura E, Miyazaki S, Horie K, Fukuda T, Hidema S, Koike M, Itakura A, Takeda S, Nishimori K. Oxytocin induced labor causes region and sex-specific transient oligodendrocyte cell death in neonatal mouse brain. J Obstet Gynaecol Res 2019; 46:66-78. [PMID: 31746074 DOI: 10.1111/jog.14149] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 10/07/2019] [Indexed: 12/16/2022]
Abstract
AIM Previous reports showed associations between oxytocin induced labor and mental disorders in offspring. However, those reports are restricted in epidemiological analyses and its mechanism remains unclear. In this study, we hypothesized that induced labor directly causes brain damage in newborns and results in the development of mental disorders. Therefore we aimed to investigate this hypothesis with animal model. METHODS The animal model of induced labor was established by subcutaneous oxytocin administration to term-pregnant C57BL/6J mice. We investigated the neonatal brain damage with evaluating immediate early gene expression (c-Fos, c-Jun and JunB) by quantitative polymerase reaction and TdT-mediated dUTP nick end labeling staining. To investigate the injured brain cell types, we performed double-immunostaining with TdT-mediated dUTP nick end labeling staining and each brain component specific protein, such as Oligo2, NeuN, GFAP and Iba1. RESULTS Brain damage during induced labor led to cell death in specific brain regions, which are implicated in mental disorders, in only male offspring at P0. Furthermore, oligodendrocyte precursors were selectively vulnerable compared to the other cell types. This oligodendrocyte-specific impairment during the perinatal period led to an increased numbers of Olig2-positive cells at P5. Expression levels of oxytocin and Oxtr in the fetal brain were not affected by the oxytocin administered to mothers during induced labor. CONCLUSION Oligodendrocyte cell death in specific brain regions, which was unrelated to the oxytocin itself, was caused by induced labor in only male offspring. This may be an underlying mechanism explaining the human epidemiological data suggesting an association between induced labor and mental disorders.
Collapse
Affiliation(s)
- Takashi Hirayama
- Graduate School of Agricultural Science, Tohoku University, Sendai, Japan.,Department of Obstetrics and Gynecology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Yuichi Hiraoka
- Graduate School of Agricultural Science, Tohoku University, Sendai, Japan.,Laboratory for Molecular Neuroscience, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Eri Kitamura
- Graduate School of Agricultural Science, Tohoku University, Sendai, Japan.,Department of Cell Biology and Neuroscience, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Shinji Miyazaki
- Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Kengo Horie
- Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Tomokazu Fukuda
- Laboratory of Cell Engineering and Molecular Genetics, Iwate University Faculty of Science and Engineering, Morioka, Japan
| | - Shizu Hidema
- Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Masato Koike
- Department of Cell Biology and Neuroscience, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Atsuo Itakura
- Department of Obstetrics and Gynecology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Satoru Takeda
- Department of Obstetrics and Gynecology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | | |
Collapse
|
128
|
Hunt BAE, Scratch SE, Mossad SI, Emami Z, Taylor MJ, Dunkley BT. Disrupted Visual Cortex Neurophysiology Following Very Preterm Birth. BIOLOGICAL PSYCHIATRY: COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2019; 5:951-960. [PMID: 31706907 DOI: 10.1016/j.bpsc.2019.08.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 08/22/2019] [Accepted: 08/25/2019] [Indexed: 11/28/2022]
Abstract
BACKGROUND Visual regions develop rapidly in utero and throughout early childhood, but very preterm (VPT) birth can disrupt the typical maturation of primary cortices, with VPT children exhibiting mild visual impairments in early life and throughout development. This is thought to be due to dysfunctional maturation of occipital cortices. A way to readily index brain function is to examine neural oscillations; these mechanisms play a central role in the modeling and pruning of connections, providing an intrinsic temporal structure that refines the precise alignment of spiking, processing information in the brain, and coordinating networks. METHODS Using magnetoencephalography, we examined regional oscillatory patterns and functional coupling in VPT and full-term children. Five minutes of eyes-open resting-state data were acquired from 27 VPT and 32 full-term children at 8 years of age. RESULTS As hypothesized, the VPT group, when compared with control children, had elevated theta-band power, while alpha amplitude envelope coupling, a marker of connectivity, was found to be decreased. CONCLUSIONS These results support the hypothesis of spectral slowing in VPT children and more broadly suggest that the developmental arc of visual neurophysiology is disrupted by VPT birth. We conclude that these deficits underlie difficulties in complex visual perceptual processing evident during childhood and beyond.
Collapse
Affiliation(s)
- Benjamin A E Hunt
- Department of Diagnostic Imaging, The Hospital for Sick Children, Toronto, Ontario, Canada; Neurosciences & Mental Health Program, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Shannon E Scratch
- Holland Bloorview Rehabilitation Hospital, Toronto, Ontario, Canada; Bloorview Research Institute, Toronto, Ontario, Canada; Department of Paediatrics, University of Toronto, Toronto, Ontario, Canada; Rehabilitation Sciences Institute, University of Toronto, Toronto, Ontario, Canada
| | - Sarah I Mossad
- Department of Diagnostic Imaging, The Hospital for Sick Children, Toronto, Ontario, Canada; Neurosciences & Mental Health Program, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada; Department of Psychology, University of Toronto, Toronto, Ontario, Canada
| | - Zahra Emami
- Department of Diagnostic Imaging, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Margot J Taylor
- Department of Diagnostic Imaging, The Hospital for Sick Children, Toronto, Ontario, Canada; Neurosciences & Mental Health Program, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada; Department of Medical Imaging, University of Toronto, Toronto, Ontario, Canada; Department of Psychology, University of Toronto, Toronto, Ontario, Canada
| | - Benjamin T Dunkley
- Department of Diagnostic Imaging, The Hospital for Sick Children, Toronto, Ontario, Canada; Neurosciences & Mental Health Program, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada; Department of Medical Imaging, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
129
|
Liu S, Jin R, Xiao AY, Zhong W, Li G. Inhibition of CD147 improves oligodendrogenesis and promotes white matter integrity and functional recovery in mice after ischemic stroke. Brain Behav Immun 2019; 82:13-24. [PMID: 31356925 PMCID: PMC6800638 DOI: 10.1016/j.bbi.2019.07.027] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 07/15/2019] [Accepted: 07/25/2019] [Indexed: 01/26/2023] Open
Abstract
White matter damage is an important contributor to long-term neurological deficit after stroke. Our previous study has shown that inhibition of CD147 ameliorates acute ischemic stroke in mice. In this study, we aimed to investigate whether inhibition of CD147 promotes white matter repair and long-term functional recovery after ischemic stroke.Male adult C57BL/6 mice were subjected to transient (1-h) middle cerebral artery occlusion (tMCAO). Anti-CD147 function-blocking antibody (αCD147) was injected intravenously once daily for 3 days beginning 4 h after onset of ischemia. Sensorimotor and cognitive functions were evaluated up to 28 days after stroke. We found that αCD147 treatment not only prevented neuronal and oligodendrocyte cell death in the acute phase, but also profoundly protected white matter integrity and reduced brain atrophy and tissue loss in the late phase, leading to improved sensorimotor and cognitive functions for at least 28 days after stroke. Mechanistically, we found that αCD147 treatment increased the number of proliferating NG2(+)/PDGFRα(+) oligodendrocyte precursor cells (OPCs) and newly generated mature APC(+)/Sox10(+) oligodendrocytes after stroke, possibly through upregulation of SDF-1/CXCR4 axis in OPCs. In conclusion, inhibition of CD147 promotes long-term functional recovery after stroke, at least in part, by enhancing oligodendrogenesis and white matter repair.
Collapse
Affiliation(s)
- Shan Liu
- Department of Neurosurgery, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Rong Jin
- Department of Neurosurgery, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Adam Y Xiao
- The Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA 71103, USA
| | - Wei Zhong
- Department of Neurosurgery, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Guohong Li
- From the Department of Neurosurgery, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA.
| |
Collapse
|
130
|
McNamara NB, Miron VE. Microglia in developing white matter and perinatal brain injury. Neurosci Lett 2019; 714:134539. [PMID: 31614181 DOI: 10.1016/j.neulet.2019.134539] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 10/04/2019] [Accepted: 10/07/2019] [Indexed: 12/13/2022]
Abstract
Perinatal brain injury (PBI) to the developing white matter results in hypomyelination of axons and can cause long-term motor and cognitive deficits e.g. cerebral palsy. There are currently no approved therapies aimed at repairing the white matter following insult, underscoring the need to investigate the mechanisms underlying the pathogenesis of PBI. Microglia have been strongly implicated, but their function and heterogeneity in this context remain poorly understood, posing a barrier to the development of microglia-targeted therapies for white matter repair following PBI. In this review, we discuss the roles of microglia in normal white matter development and in PBI, and potential drug strategies to influence microglial responses in this setting.
Collapse
Affiliation(s)
- Niamh B McNamara
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Veronique E Miron
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom.
| |
Collapse
|
131
|
Borshchev YY, Uspensky YP, Galagudza MM. Pathogenetic pathways of cognitive dysfunction and dementia in metabolic syndrome. Life Sci 2019; 237:116932. [PMID: 31606384 DOI: 10.1016/j.lfs.2019.116932] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 09/27/2019] [Accepted: 10/02/2019] [Indexed: 12/13/2022]
Abstract
The prevalence of dementia worldwide is growing at an alarming rate. A number of studies and meta-analyses have provided evidence for increased risk of dementia in patients with metabolic syndrome (MS) as compared to persons without MS. However, there are some reports demonstrating a lack of association between MS and increased dementia risk. In this review, taking into account the potential role of individual MS components in the pathogenesis of MS-related cognitive dysfunction, we considered the underlying mechanisms in arterial hypertension, diabetes mellitus, dyslipidemia, and obesity. The pathogenesis of dementia in MS is multifactorial, involving both vascular injury and non-ischemic neuronal death due to neurodegeneration. Neurodegenerative and ischemic lesions do not simply coexist in the brain due to independent evolution, but rather exacerbate each other, leading to more severe consequences for cognition than would either pathology alone. In addition to universal mechanisms of cognitive dysfunction shared by all MS components, other pathogenetic pathways leading to cognitive deficits and dementia, which are specific for each component, also play a role. Examples of such component-specific pathogenetic pathways include central insulin resistance and hypoglycemia in diabetes, neuroinflammation and adipokine imbalance in obesity, as well as arteriolosclerosis and lipohyalinosis in arterial hypertension. A more detailed understanding of cognitive disorders based on the recognition of underlying molecular mechanisms will aid in the development of new methods for prevention and treatment of devastating cognitive problems in MS.
Collapse
Affiliation(s)
- Yury Yu Borshchev
- Institute of Experimental Medicine, Almazov National Medical Research Center, Saint Petersburg, Russian Federation
| | - Yury P Uspensky
- Department of Faculty Therapy, Saint Petersburg State Pediatric Medical University, Saint Petersburg, Russian Federation
| | - Michael M Galagudza
- Laboratory of Digital and Display Holography, ITMO University, Russian Federation, Saint Petersburg, Russian Federation.
| |
Collapse
|
132
|
Zhao H, Gao XY, Liu ZH, Lin JW, Wang SP, Wang DX, Zhang YB. Effects of the transcription factor Olig1 on the differentiation and remyelination of oligodendrocyte precursor cells after focal cerebral ischemia in rats. Mol Med Rep 2019; 20:4603-4611. [PMID: 31702031 PMCID: PMC6797933 DOI: 10.3892/mmr.2019.10713] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Accepted: 07/22/2019] [Indexed: 12/28/2022] Open
Abstract
The differentiation and maturation of oligodendrocyte precursor cells (OPCs) is important for remyelination in the central nervous system. Nevertheless, this process is often limited and incomplete in ischemic injury. Oligodendrocyte transcription factor 1 (Olig1) is important for the maturation of OPCs and the repair of demyelinated lesions. However, how Olig1 modulates the development of OPCs or the remyelination associated with ischemic injury remains unclear. The present study aimed to examine alterations in OPCs, and the expression of myelin and Olig1, at different time-points after focal cerebral ischemia using immunohistochemistry and western blot techniques to elucidate the role of Olig1 in the maturation of OPCs and remyelination. The present results showed that the expression of Olig1 significantly decreased at 1 day after middle cerebral artery occlusion (MCAO) and returned to normal levels from day 3 to 28. Additionally, Olig1 was found to translocate into the nucleus following ischemia in the brain. The number of OPCs in the ischemic striatum significantly declined at days 1 and 3 following MCAO, and increased at days 7, 14 and 28 compared with the control. The expression of myelin basic protein, a marker of mature oligodendrocytes and myelin, gradually decreased from day 1 to 7 after ischemia and recovered at day 14 and 28; however, the levels were lower than those in the control group. The present results indicated that the restored normal level of Olig1 following ischemia may play an important role in the maturation of OPCs through its translocation into the nucleus, where it may promote the growth and development of myelin under pathological conditions. However, this endogenous recovery mechanism fails to fully repair the demyelinated lesion. The data of the present study may help clinicians understand the expression pattern of Olig1 and its potential role in endogenous remyelination after ischemia, which may have implications for the treatment of diseases that lead to demyelination.
Collapse
Affiliation(s)
- Hong Zhao
- Department of Neurology, Dalian Municipal Central Hospital, Dalian, Liaoning 116033, P.R. China
| | - Xiao-Yu Gao
- Department of Neurology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, P.R. China
| | - Zan-Hua Liu
- Department of Neurology, Nanjing Gaochun People's Hospital, Nanjing, Jiangsu 211300, P.R. China
| | - Jian-Wen Lin
- Department of Neurology, Dalian Municipal Central Hospital, Dalian, Liaoning 116033, P.R. China
| | - Su-Ping Wang
- Department of Neurology, Dalian Municipal Central Hospital, Dalian, Liaoning 116033, P.R. China
| | - De-Xin Wang
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P.R. China
| | - Yong-Bo Zhang
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P.R. China
| |
Collapse
|
133
|
Ceprián M, Vargas C, García-Toscano L, Penna F, Jiménez-Sánchez L, Achicallende S, Elezgarai I, Grandes P, Hind W, Pazos MR, Martínez-Orgado J. Cannabidiol Administration Prevents Hypoxia-Ischemia-Induced Hypomyelination in Newborn Rats. Front Pharmacol 2019; 10:1131. [PMID: 31611802 PMCID: PMC6775595 DOI: 10.3389/fphar.2019.01131] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 08/30/2019] [Indexed: 01/09/2023] Open
Abstract
Neonatal hypoxia-ischemia (HI) is a risk factor for myelination disturbances, a key factor for cerebral palsy. Cannabidiol (CBD) protects neurons and glial cells after HI insult in newborn animals. We hereby aimed to study CBD’s effects on long-lasting HI-induced myelination deficits in newborn rats. Thus, P7 Wistar rats received s.c. vehicle (HV) or cannabidiol (HC) after HI brain damage (left carotid artery electrocoagulation plus 10% O2 for 112 min). Controls were non-HI pups. At P37, neurobehavioral tests were performed and immunohistochemistry [quantifying mature oligodendrocyte (mOL) populations and myelin basic protein (MBP) density] and electron microscopy (determining axon number, size, and myelin thickness) studies were conducted in cortex (CX) and white matter (WM). Expression of brain-derived neurotrophic factor (BDNF) and glial-derived neurotrophic factor (GDNF) were analyzed by western blot at P14. HI reduced mOL or MBP in CX but not in WM. In both CX and WM, axon density and myelin thickness were reduced. MBP impairment correlated with functional deficits. CBD administration resulted in normal function associated with normal mOL and MBP, as well as normal axon density and myelin thickness in all areas. CBD’s effects were not associated with increased BDNF or GDNF expression. In conclusion, HI injury in newborn rats resulted in long-lasting myelination disturbance, associated with functional impairment. CBD treatment preserved function and myelination, likely as a part of a general neuroprotective effect.
Collapse
Affiliation(s)
- María Ceprián
- Department of Experimental Medicine, Health Research Institute Puerta de Hierro Majadahonda, Madrid, Spain.,Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense, Madrid, Spain
| | - Carlos Vargas
- Division of Neonatology, Hospital Clínico San Carlos - IdISSC, Madrid, Spain
| | - Laura García-Toscano
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense, Madrid, Spain.,CIBER de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Federica Penna
- Department of DBSV, Laboratory of Neuropsychopharmacology, University of Insubria, Varese, Italy
| | - Laura Jiménez-Sánchez
- Department of Experimental Medicine, Health Research Institute Puerta de Hierro Majadahonda, Madrid, Spain
| | - Svein Achicallende
- School of Medicine and Nursery, Universidad del País Vasco, Bilbao, Spain
| | - Izaskun Elezgarai
- School of Medicine and Nursery, Universidad del País Vasco, Bilbao, Spain
| | - Pedro Grandes
- School of Medicine and Nursery, Universidad del País Vasco, Bilbao, Spain
| | | | - M Ruth Pazos
- Department of Experimental Medicine, Health Research Institute Puerta de Hierro Majadahonda, Madrid, Spain.,Laboratorio de Apoyo a la Investigación, Hospital Universitario Fundación Alcorcón, Madrid, Spain
| | - José Martínez-Orgado
- Department of Experimental Medicine, Health Research Institute Puerta de Hierro Majadahonda, Madrid, Spain.,Division of Neonatology, Hospital Clínico San Carlos - IdISSC, Madrid, Spain
| |
Collapse
|
134
|
Edwards BA, Nava-Guerra L, Kemp JS, Carroll JL, Khoo MC, Sands SA, Terrill PI, Landry SA, Amin RS. Assessing ventilatory instability using the response to spontaneous sighs during sleep in preterm infants. Sleep 2019; 41:5077835. [PMID: 30137560 DOI: 10.1093/sleep/zsy161] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Indexed: 12/15/2022] Open
Abstract
Study Objectives Periodic breathing (PB) is common in newborns and is an obvious manifestation of ventilatory control instability. However, many infants without PB may still have important underlying ventilatory control instabilities that go unnoticed using standard clinical monitoring. Methods to detect infants with "subclinical" ventilatory control instability are therefore required. The current study aimed to assess the degree of ventilatory control instability using simple bedside recordings in preterm infants. Methods Respiratory inductance plethysmography (RIP) recordings were analyzed from ~20 minutes of quiet sleep in 20 preterm infants at 36 weeks post-menstrual age (median [range]: 36 [34-40]). The percentage time spent in PB was also calculated for each infant (%PB). Spontaneous sighs were identified and breath-by-breath measurements of (uncalibrated) ventilation were derived from RIP traces. Loop gain (LG, a measure of ventilatory control instability) was calculated by fitting a simple ventilatory control model (gain, time-constant, delay) to the post-sigh ventilatory pattern. For comparison, periodic inter-breath variability was also quantified using power spectral analysis (ventilatory oscillation magnitude index [VOMI]). Results %PB was strongly associated with LG (r2 = 0.77, p < 0.001) and moderately with the VOMI (r2 = 0.21, p = 0.047). LG (0.52 ± 0.05 vs. 0.30 ± 0.03; p = 0.0025) and the VOMI (-8.2 ± 1.1 dB vs. -11.8 ± 0.9 dB; p = 0.026) were both significantly higher in infants that displayed PB vs. those without. Conclusions LG and VOMI determined from the ventilatory responses to spontaneous sighs can provide a practical approach to assessing ventilatory control instability in preterm infants. Such simple techniques may help identify infants at particular risk for ventilatory instabilities with concomitant hypoxemia and its associated consequences.
Collapse
Affiliation(s)
- Bradley A Edwards
- Sleep and Circadian Medicine Laboratory, Department of Physiology, Monash University, Melbourne, Australia.,School of Psychological Sciences and Monash Institute of Cognitive and Clinical Neurosciences, Monash University, Melbourne, Australia.,Division of Sleep and Circadian Disorders, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Leonardo Nava-Guerra
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA
| | - James S Kemp
- Division of Allergy, Immunology and Pulmonary Medicine, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO
| | - John L Carroll
- Division of Pediatric Pulmonary and Sleep Medicine, Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR
| | - Michael C Khoo
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA
| | - Scott A Sands
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Philip I Terrill
- School of Information Technology and Electrical Engineering, University of Queensland, Brisbane, Australia
| | - Shane A Landry
- Sleep and Circadian Medicine Laboratory, Department of Physiology, Monash University, Melbourne, Australia.,School of Psychological Sciences and Monash Institute of Cognitive and Clinical Neurosciences, Monash University, Melbourne, Australia
| | - Raouf S Amin
- Division of Pulmonary Medicine, Department of Pediatrics, Cincinnati Children Hospital Medical Center, Cincinnati, OH
| |
Collapse
|
135
|
Gonzalo H, Nogueras L, Gil-Sánchez A, Hervás JV, Valcheva P, González-Mingot C, Martin-Gari M, Canudes M, Peralta S, Solana MJ, Pamplona R, Portero-Otin M, Boada J, Serrano JCE, Brieva L. Impairment of Mitochondrial Redox Status in Peripheral Lymphocytes of Multiple Sclerosis Patients. Front Neurosci 2019; 13:938. [PMID: 31551694 PMCID: PMC6738270 DOI: 10.3389/fnins.2019.00938] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 08/21/2019] [Indexed: 11/13/2022] Open
Abstract
Literature suggests that oxidative stress (OS) may be involved in the pathogenesis of multiple sclerosis (MS), in which the immune system is known to play a key role. However, to date, the OS in peripheral lymphocytes and its contribution to the disease remain unknown. The aim of the present study was to explore the influence of OS in peripheral lymphocytes of MS patients. To that end, a cross-sectional, observational pilot study was conducted [n = 58: 34 MS and 24 healthy subjects (control group)]. We have measured superoxide production and protein mitochondrial complex levels in peripheral blood mononuclear cells (PBMCs) isolated from MS patients and control. Lactate levels and the antioxidant capacity were determined in plasma. We adjusted the comparisons between study groups by age, sex and cell count according to case. Results demonstrated that PBMCs, specifically T cells, from MS patients exhibited significantly increased superoxide anion production compared to control group (p = 0.027 and p = 0.041, respectively). Increased superoxide production in PBMCs was maintained after the adjustment (p = 0.044). Regarding mitochondrial proteins, we observe a significant decrease in the representative protein content of the mitochondrial respiratory chain complexes I-V in PBMCs of MS patients (p = 0.002, p = 0.037, p = 0.03, p = 0.044, and p = 0.051, respectively), which was maintained for complexes I, III, and V after the adjustment (p = 0.026; p = 0.033; p = 0.033, respectively). In MS patients, a trend toward increased plasma lactate concentration was detected [8.04 mg lactate/dL (5.25, 9.49) in the control group, 11.36 mg lactate/dL (5.41, 14.81) in MS patients] that was statistically significant after the adjustment (p = 0.013). This might be indicative of compromised mitochondrial function. Finally, antioxidant capacity was also decreased in plasma from MS patients, both before (p = 0.027) and after adjusting for sex and age (p = 0.006). Our findings demonstrate that PBMCs of MS patients show impaired mitochondrial redox status and deficient antioxidant capacity. These results demonstrate for the first time the existence of mitochondrial alterations in the cells immune cells of MS patients already at the peripheral level.
Collapse
Affiliation(s)
- Hugo Gonzalo
- Institut de Recerca Biomèdica de Lleida, Lleida, Spain.,Clinical University Hospital of Valladolid (HCUV), Department of Research and Innovation, SACYL/IECSCYL, Valladolid, Spain
| | - Lara Nogueras
- Universitat de Lleida, Departament de Medicina Experimental, Lleida, Spain
| | | | | | | | | | | | - Marc Canudes
- Institut de Recerca Biomèdica de Lleida, Lleida, Spain
| | | | | | - Reinald Pamplona
- Universitat de Lleida, Departament de Medicina Experimental, Lleida, Spain
| | | | - Jordi Boada
- Universitat de Lleida, Departament de Medicina Experimental, Lleida, Spain
| | | | - Luis Brieva
- Hospital Universitario Arnau de Vilanova, Lleida, Spain
| |
Collapse
|
136
|
Cavarsan CF, Gorassini MA, Quinlan KA. Animal models of developmental motor disorders: parallels to human motor dysfunction in cerebral palsy. J Neurophysiol 2019; 122:1238-1253. [PMID: 31411933 PMCID: PMC6766736 DOI: 10.1152/jn.00233.2019] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 08/07/2019] [Accepted: 08/08/2019] [Indexed: 12/12/2022] Open
Abstract
Cerebral palsy (CP) is the most common motor disability in children. Much of the previous research on CP has focused on reducing the severity of brain injuries, whereas very few researchers have investigated the cause and amelioration of motor symptoms. This research focus has had an impact on the choice of animal models. Many of the commonly used animal models do not display a prominent CP-like motor phenotype. In general, rodent models show anatomically severe injuries in the central nervous system (CNS) in response to insults associated with CP, including hypoxia, ischemia, and neuroinflammation. Unfortunately, most rodent models do not display a prominent motor phenotype that includes the hallmarks of spasticity (muscle stiffness and hyperreflexia) and weakness. To study motor dysfunction related to developmental injuries, a larger animal model is needed, such as rabbit, pig, or nonhuman primate. In this work, we describe and compare various animal models of CP and their potential for translation to the human condition.
Collapse
Affiliation(s)
- Clarissa F Cavarsan
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, Rhode Island
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island
| | - Monica A Gorassini
- Department of Biomedical Engineering, University of Alberta, Edmonton, Alberta, Canada
| | - Katharina A Quinlan
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, Rhode Island
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island
| |
Collapse
|
137
|
Shin SH, Kim EK, Lee KY, Kim HS. TNF-α antagonist attenuates systemic lipopolysaccharide-induced brain white matter injury in neonatal rats. BMC Neurosci 2019; 20:45. [PMID: 31470812 PMCID: PMC6716877 DOI: 10.1186/s12868-019-0529-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 08/26/2019] [Indexed: 12/04/2022] Open
Abstract
Background Systemic inflammation is an important risk factor for neurodevelopmental impairments in preterm infants. Premyelinating oligodendrocytes are main building blocks of white matter in preterm infants and vulnerable to oxidative stress and excitotoxic stress. Tumour necrosis factor-α (TNF-α) plays important roles in systemic inflammation and local inflammation leading to apoptosis of premyelinating oligodendrocytes and white matter injury (WMI) in brain tissue. This study was conducted to investigate whether etanercept, a TNF-α antagonist, could attenuate systemic lipopolysaccharide (LPS)-induced WMI in the immature brain. Results We found that intraperitoneal LPS administration caused systemic and local inflammation in brain tissue. Subsequent etanercept treatment significantly attenuated LPS-induced inflammation in brain tissue as well as in systemic circulation. Intraperitoneal LPS also induced microgliosis and astrocytosis in the cingulum and etanercept treatment reduced LPS-induced microgliosis and astrocytosis. Additionally, systemic LPS-induced apoptosis of oligodendrocyte precursor cells was observed, which was lessened by etanercept treatment. The concentration of etanercept in the CSF was higher when it was administrated with LPS than when administrated with a vehicle. Conclusions It appears that etanercept reduce WMI in the neonatal rat brain via attenuation of systemic and local inflammation. This study provides preclinical data suggesting etanercept-mediated modulation of inflammation as a promising approach to reduce WMI caused by sepsis or necrotizing enterocolitis in preterm infants.
Collapse
Affiliation(s)
- Seung Han Shin
- Department of Pediatrics, Seoul National University College of Medicine, Seoul National University Children's Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 110-769, South Korea
| | - Ee-Kyung Kim
- Department of Pediatrics, Seoul National University College of Medicine, Seoul National University Children's Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 110-769, South Korea.
| | - Kyung-Yup Lee
- Department of Pediatrics, Seoul National University College of Medicine, Seoul National University Children's Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 110-769, South Korea
| | - Han-Suk Kim
- Department of Pediatrics, Seoul National University College of Medicine, Seoul National University Children's Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 110-769, South Korea
| |
Collapse
|
138
|
Jensen BK, Roth LM, Grinspan JB, Jordan-Sciutto KL. White matter loss and oligodendrocyte dysfunction in HIV: A consequence of the infection, the antiretroviral therapy or both? Brain Res 2019; 1724:146397. [PMID: 31442414 DOI: 10.1016/j.brainres.2019.146397] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 08/08/2019] [Accepted: 08/19/2019] [Indexed: 01/13/2023]
Abstract
While the severe cognitive effects of HIV-associated dementia have been reduced by combined antiretroviral therapy (cART), nearly half of HIV-positive (HIV+) patients still suffer from some form of HIV-Associated Neurocognitive Disorders (HAND). While frank neuronal loss has been dramatically reduced in HAND patients, white matter loss, including dramatic thinning of the corpus callosum, and loss of volume and structural integrity of myelin persists despite viral control by cART. It remains unclear whether changes in white matter underlie the clinical manifestation seen in patients or whether they are the result of persistent viral reservoirs, remnant damage from the acute infection, the antiretroviral compounds used to treat HIV, secondary effects due to peripheral toxicities or other associated comorbid conditions. Both HIV infection itself and its treatment with antiretroviral drugs can induce metabolic syndrome, lipodystrophy, atherosclerosis and peripheral neuropathies by increased oxidative stress, induction of the unfolded protein response and dysregulation of lipid metabolism. These virally and/or cART-induced processes can also cause myelin loss in the CNS. This review aims to highlight existing data on the contribution of white matter damage to HAND and explore the mechanisms by which HIV infection and its treatment contribute to persistence of white matter changes in people living with HIV currently on cART.
Collapse
Affiliation(s)
- Brigid K Jensen
- Vickie and Jack Farber Institute for Neuroscience, Jefferson Weinberg ALS Center, Thomas Jefferson University, United States; Department of Neurology, The Children's Hospital of Philadelphia, United States; Department of Pathology, School of Dental Medicine, University of Pennsylvania, United States
| | - Lindsay M Roth
- Department of Neurology, The Children's Hospital of Philadelphia, United States; Department of Pathology, School of Dental Medicine, University of Pennsylvania, United States
| | - Judith B Grinspan
- Department of Pathology, School of Dental Medicine, University of Pennsylvania, United States
| | | |
Collapse
|
139
|
Protective effects of delayed intraventricular TLR7 agonist administration on cerebral white and gray matter following asphyxia in the preterm fetal sheep. Sci Rep 2019; 9:9562. [PMID: 31267031 PMCID: PMC6606639 DOI: 10.1038/s41598-019-45872-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 06/13/2019] [Indexed: 01/08/2023] Open
Abstract
Preterm brain injury is highly associated with inflammation, which is likely related in part to sterile responses to hypoxia-ischemia. We have recently shown that neuroprotection with inflammatory pre-conditioning in the immature brain is associated with induction of toll-like receptor 7 (TLR7). We therefore tested the hypothesis that central administration of a synthetic TLR7 agonist, gardiquimod (GDQ), after severe hypoxia-ischemia in preterm-equivalent fetal sheep would improve white and gray matter recovery. Fetal sheep at 0.7 of gestation received sham asphyxia or asphyxia induced by umbilical cord occlusion for 25 minutes, followed by a continuous intracerebroventricular infusion of GDQ or vehicle from 1 to 4 hours (total dose 1.8 mg/kg). Sheep were killed 72 hours after asphyxia for histology. GDQ significantly improved survival of immature and mature oligodendrocytes (2′,3′-cyclic-nucleotide 3′-phosphodiesterase, CNPase) and total oligodendrocytes (oligodendrocyte transcription factor 2, Olig-2) within the periventricular and intragyral white matter. There were reduced numbers of cells showing cleaved caspase-3 positive apoptosis and astrogliosis (glial fibrillary acidic protein, GFAP) in both white matter regions. Neuronal survival was increased in the dentate gyrus, caudate and medial thalamic nucleus. Central infusion of GDQ was associated with a robust increase in fetal plasma concentrations of the anti-inflammatory cytokines, interferon-β (IFN-β) and interleukin-10 (IL-10), with no significant change in the concentration of the pro-inflammatory cytokine, tumor necrosis factor-α (TNF-α). In conclusion, delayed administration of the TLR7 agonist, GDQ, after severe hypoxia-ischemia in the developing brain markedly ameliorated white and gray matter damage, in association with upregulation of anti-inflammatory cytokines. These data strongly support the hypothesis that modulation of secondary inflammation may be a viable therapeutic target for injury of the preterm brain.
Collapse
|
140
|
Ceprian M, Fulton D. Glial Cell AMPA Receptors in Nervous System Health, Injury and Disease. Int J Mol Sci 2019; 20:E2450. [PMID: 31108947 PMCID: PMC6566241 DOI: 10.3390/ijms20102450] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 04/11/2019] [Accepted: 04/22/2019] [Indexed: 12/16/2022] Open
Abstract
Glia form a central component of the nervous system whose varied activities sustain an environment that is optimised for healthy development and neuronal function. Alpha-amino-3-hydroxy-5-methyl-4-isoxazole (AMPA)-type glutamate receptors (AMPAR) are a central mediator of glutamatergic excitatory synaptic transmission, yet they are also expressed in a wide range of glial cells where they influence a variety of important cellular functions. AMPAR enable glial cells to sense the activity of neighbouring axons and synapses, and as such many aspects of glial cell development and function are influenced by the activity of neural circuits. However, these AMPAR also render glia sensitive to elevations of the extracellular concentration of glutamate, which are associated with a broad range of pathological conditions. Excessive activation of AMPAR under these conditions may induce excitotoxic injury in glial cells, and trigger pathophysiological responses threatening other neural cells and amplifying ongoing disease processes. The aim of this review is to gather information on AMPAR function from across the broad diversity of glial cells, identify their contribution to pathophysiological processes, and highlight new areas of research whose progress may increase our understanding of nervous system dysfunction and disease.
Collapse
Affiliation(s)
- Maria Ceprian
- Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain.
- Departamento de Bioquímica y Biología Molecular, CIBERNED, IRICYS. Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain.
| | - Daniel Fulton
- Neuroscience and Ophthalmology Research Group, Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK.
| |
Collapse
|
141
|
Shaw JC, Berry MJ, Dyson RM, Crombie GK, Hirst JJ, Palliser HK. Reduced Neurosteroid Exposure Following Preterm Birth and Its' Contribution to Neurological Impairment: A Novel Avenue for Preventative Therapies. Front Physiol 2019; 10:599. [PMID: 31156466 PMCID: PMC6529563 DOI: 10.3389/fphys.2019.00599] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 04/26/2019] [Indexed: 12/21/2022] Open
Abstract
Children born preterm are at an increased risk of developing cognitive problems and neuro-behavioral disorders such as attention deficit hyperactivity disorder (ADHD) and anxiety. Whilst neonates born at all gestational ages, even at term, can experience poor cognitive outcomes due to birth-complications such as birth asphyxia, it is becoming widely known that children born preterm in particular are at significant risk for learning difficulties with an increased utilization of special education resources, when compared to their healthy term-born peers. Additionally, those born preterm have evidence of altered cerebral myelination with reductions in white matter volumes of the frontal cortex, hippocampus and cerebellum evident on magnetic resonance imaging (MRI). This disruption to myelination may underlie some of the pathophysiology of preterm-associated brain injury. Compared to a fetus of the same post-conceptional age, the preterm newborn loses access to in utero factors that support and promote healthy brain development. Furthermore, the preterm ex utero environment is hostile to the developing brain with a myriad of environmental, biochemical and excitotoxic stressors. Allopregnanolone is a key neuroprotective fetal neurosteroid which has promyelinating effects in the developing brain. Preterm birth leads to an abrupt loss of the protective effects of allopregnanolone, with a dramatic drop in allopregnanolone concentrations in the preterm neonatal brain compared to the fetal brain. This occurs in conjunction with reduced myelination of the hippocampus, subcortical white matter and cerebellum; thus, damage to neurons, astrocytes and especially oligodendrocytes of the developing nervous system can occur in the vulnerable developmental window prior to term as a consequence reduced allopregnanolone. In an effort to prevent preterm-associated brain injury a number of therapies have been considered, but to date, other than antenatal magnesium sulfate and corticosteroid therapy, none have become part of standard clinical care for vulnerable infants. Therefore, there remains an urgent need for improved therapeutic options to prevent brain injury in preterm neonates. The actions of the placentally derived neurosteroid allopregnanolone on GABAA receptor signaling has a major role in late gestation neurodevelopment. The early loss of this intrauterine neurotrophic support following preterm birth may be pivotal to development of neurodevelopmental morbidity. Thus, restoring the in utero neurosteroid environment for preterm neonates may represent a new and clinically feasible treatment option for promoting better trajectories of myelination and brain development, and therefore reducing neurodevelopmental disorders in children born preterm.
Collapse
Affiliation(s)
- Julia C. Shaw
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, NSW, Australia
- Mothers and Babies Research Centre, Hunter Medical Research Institute, University of Newcastle, Newcastle, NSW, Australia
| | - Mary J. Berry
- Department of Paediatrics and Child Health, University of Otago, Wellington, Wellington, New Zealand
- Centre for Translational Physiology, University of Otago, Wellington, Wellington, New Zealand
| | - Rebecca M. Dyson
- Department of Paediatrics and Child Health, University of Otago, Wellington, Wellington, New Zealand
- Centre for Translational Physiology, University of Otago, Wellington, Wellington, New Zealand
| | - Gabrielle K. Crombie
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, NSW, Australia
- Mothers and Babies Research Centre, Hunter Medical Research Institute, University of Newcastle, Newcastle, NSW, Australia
| | - Jonathan J. Hirst
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, NSW, Australia
- Mothers and Babies Research Centre, Hunter Medical Research Institute, University of Newcastle, Newcastle, NSW, Australia
| | - Hannah K. Palliser
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, NSW, Australia
- Mothers and Babies Research Centre, Hunter Medical Research Institute, University of Newcastle, Newcastle, NSW, Australia
| |
Collapse
|
142
|
Goussakov I, Synowiec S, Yarnykh V, Drobyshevsky A. Immediate and delayed decrease of long term potentiation and memory deficits after neonatal intermittent hypoxia. Int J Dev Neurosci 2019; 74:27-37. [PMID: 30858028 PMCID: PMC6461389 DOI: 10.1016/j.ijdevneu.2019.03.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 03/07/2019] [Accepted: 03/07/2019] [Indexed: 11/25/2022] Open
Abstract
Apnea of prematurity is a common clinical condition that occurs in premature infants and results in intermittent hypoxia (IH) to brain and other organs. While short episodes of apnea are considered of no clinical significance, prolonged apnea with bradycardia and large oxygen desaturation is associated with adverse neurological and cognitive outcome. The mechanisms of cognitive deficits in IH are poorly understood. We hypothesized that brief but multiple episodes of severe oxygen desaturation accompanied by bradycardia may affect early and late synaptic plasticity and produce long-term cognitive deficits. C57BL/6 mouse pups were exposed to IH paradigm consisting of alternating cycles of 5% oxygen for 2.5 min and room air for 5-10 min, 2 h a day from P3 to P7. Long term potentiation (LTP) of synaptic strength in response to high frequency stimulation in hippocampal slices were examined 3 days and 6 weeks after IH. LTP was decreased in IH group relative to controls at both time points. That decrease was associated with deficits in spatial memory on Morris water maze and context fear conditioning test. Hypomyelination was observed in multiple gray and white matter areas on in vivo MRI using micromolecule proton fraction and ex vivo diffusion tensor imaging. No difference in caspase labeling was found between control and IH groups. We conclude that early changes in synaptic plasticity occurring during severe episodes of neonatal IH and persisting to adulthood may represent functional and structural substrate for long term cognitive deficits.
Collapse
Affiliation(s)
- Ivan Goussakov
- Department of Pediatrics, NorthShore University HealthSystem Research Institute, 2650 Ridge Ave 60201, Evanston, IL, USA
| | - Sylvia Synowiec
- Department of Pediatrics, NorthShore University HealthSystem Research Institute, 2650 Ridge Ave 60201, Evanston, IL, USA
| | - Vasily Yarnykh
- Department of Radiology, University of Washington, 850 Republican St., Room 255 Seattle, WA, USA
| | - Alexander Drobyshevsky
- Department of Pediatrics, NorthShore University HealthSystem Research Institute, 2650 Ridge Ave 60201, Evanston, IL, USA.
| |
Collapse
|
143
|
Pregnolato S, Chakkarapani E, Isles AR, Luyt K. Glutamate Transport and Preterm Brain Injury. Front Physiol 2019; 10:417. [PMID: 31068830 PMCID: PMC6491644 DOI: 10.3389/fphys.2019.00417] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 03/27/2019] [Indexed: 12/19/2022] Open
Abstract
Preterm birth complications are the leading cause of child death worldwide and a top global health priority. Among the survivors, the risk of life-long disabilities is high, including cerebral palsy and impairment of movement, cognition, and behavior. Understanding the molecular mechanisms of preterm brain injuries is at the core of future healthcare improvements. Glutamate excitotoxicity is a key mechanism in preterm brain injury, whereby the accumulation of extracellular glutamate damages the delicate immature oligodendrocytes and neurons, leading to the typical patterns of injury seen in the periventricular white matter. Glutamate excitotoxicity is thought to be induced by an interaction between environmental triggers of injury in the perinatal period, particularly cerebral hypoxia-ischemia and infection/inflammation, and developmental and genetic vulnerabilities. To avoid extracellular build-up of glutamate, the brain relies on rapid uptake by sodium-dependent glutamate transporters. Astrocytic excitatory amino acid transporter 2 (EAAT2) is responsible for up to 95% of glutamate clearance, and several lines of evidence suggest that it is essential for brain functioning. While in the adult EAAT2 is predominantly expressed by astrocytes, EAAT2 is transiently upregulated in the immature oligodendrocytes and selected neuronal populations during mid-late gestation, at the peak time for preterm brain injury. This developmental upregulation may interact with perinatal hypoxia-ischemia and infection/inflammation and contribute to the selective vulnerability of the immature oligodendrocytes and neurons in the preterm brain. Disruption of EAAT2 may involve not only altered expression but also impaired function with reversal of transport direction. Importantly, elevated EAAT2 levels have been found in the reactive astrocytes and macrophages of human infant post-mortem brains with severe white matter injury (cystic periventricular leukomalacia), potentially suggesting an adaptive mechanism against excitotoxicity. Interestingly, EAAT2 is suppressed in animal models of acute hypoxic-ischemic brain injury at term, pointing to an important and complex role in newborn brain injuries. Enhancement of EAAT2 expression and transport function is gathering attention as a potential therapeutic approach for a variety of adult disorders and awaits exploration in the context of the preterm brain injuries.
Collapse
Affiliation(s)
- Silvia Pregnolato
- Department of Neonatal Neurology, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Elavazhagan Chakkarapani
- Department of Neonatal Neurology, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Anthony R Isles
- Behavioural Genetics Group, MRC Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Karen Luyt
- Department of Neonatal Neurology, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
144
|
Zeng Y, Wang H, Zhang L, Tang J, Shi J, Xiao D, Qu Y, Mu D. The optimal choices of animal models of white matter injury. Rev Neurosci 2019; 30:245-259. [PMID: 30379639 DOI: 10.1515/revneuro-2018-0044] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 06/16/2018] [Indexed: 12/25/2022]
Abstract
White matter injury, the most common neurological injury in preterm infants, is a major cause of chronic neurological morbidity, including cerebral palsy. Although there has been great progress in the study of the mechanism of white matter injury in newborn infants, its pathogenesis is not entirely clear, and further treatment approaches are required. Animal models are the basis of study in pathogenesis, treatment, and prognosis of white matter injury in preterm infants. Various species have been used to establish white matter injury models, including rodents, rabbits, sheep, and non-human primates. Small animal models allow cost-effective investigation of molecular and cellular mechanisms, while large animal models are particularly attractive for pathophysiological and clinical-translational studies. This review focuses on the features of commonly used white matter injury animal models, including their modelling methods, advantages, and limitations, and addresses some clinically relevant animal models that allow reproduction of the insults associated with clinical conditions that contribute to white matter injury in human infants.
Collapse
Affiliation(s)
- Yan Zeng
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China.,Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, Sichuan University, Chengdu 610041, China
| | - Huiqing Wang
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China.,Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, Sichuan University, Chengdu 610041, China
| | - Li Zhang
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China.,Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, Sichuan University, Chengdu 610041, China
| | - Jun Tang
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China.,Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, Sichuan University, Chengdu 610041, China
| | - Jing Shi
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China.,Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, Sichuan University, Chengdu 610041, China
| | - Dongqiong Xiao
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China.,Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, Sichuan University, Chengdu 610041, China
| | - Yi Qu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China.,Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, Sichuan University, Chengdu 610041, China
| | - Dezhi Mu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, No. 20, section 3, Renmin South Road, Chengdu, Sichuan 610041, China.,Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, Sichuan University, Chengdu 610041, China, Telephone: +86-28-85503226, Fax: +86-28-85559065
| |
Collapse
|
145
|
Hedderich DM, Bäuml JG, Berndt MT, Menegaux A, Scheef L, Daamen M, Zimmer C, Bartmann P, Boecker H, Wolke D, Gaser C, Sorg C. Aberrant gyrification contributes to the link between gestational age and adult IQ after premature birth. Brain 2019; 142:1255-1269. [DOI: 10.1093/brain/awz071] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 01/23/2019] [Accepted: 01/30/2019] [Indexed: 12/31/2022] Open
Affiliation(s)
- Dennis M Hedderich
- TUM-NIC Neuroimaging Center, Technische Universität München, Munich, Germany
- Department of Neuroradiology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Josef G Bäuml
- TUM-NIC Neuroimaging Center, Technische Universität München, Munich, Germany
- Department of Neuroradiology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Maria T Berndt
- TUM-NIC Neuroimaging Center, Technische Universität München, Munich, Germany
- Department of Neuroradiology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Aurore Menegaux
- TUM-NIC Neuroimaging Center, Technische Universität München, Munich, Germany
- Department of Neuroradiology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
- Graduate School of Systemic Neurosciences GSN, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Lukas Scheef
- Functional Neuroimaging Group, Department of Radiology, University Hospital Bonn, Bonn, Germany
| | - Marcel Daamen
- Functional Neuroimaging Group, Department of Radiology, University Hospital Bonn, Bonn, Germany
- Department of Neonatology, University Hospital Bonn, Bonn, Germany
| | - Claus Zimmer
- Department of Neuroradiology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Peter Bartmann
- Department of Neonatology, University Hospital Bonn, Bonn, Germany
| | - Henning Boecker
- Functional Neuroimaging Group, Department of Radiology, University Hospital Bonn, Bonn, Germany
| | - Dieter Wolke
- Department of Psychology, University of Warwick, Coventry, UK
- Warwick Medical School, University of Warwick, Coventry, UK
| | - Christian Gaser
- Department of Psychiatry and Neurology, University Hospital Jena, Jena, Germany
| | - Christian Sorg
- TUM-NIC Neuroimaging Center, Technische Universität München, Munich, Germany
- Department of Neuroradiology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
- Department of Psychiatry, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| |
Collapse
|
146
|
Rodent Models of Developmental Ischemic Stroke for Translational Research: Strengths and Weaknesses. Neural Plast 2019; 2019:5089321. [PMID: 31093271 PMCID: PMC6476045 DOI: 10.1155/2019/5089321] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 12/19/2018] [Accepted: 02/06/2019] [Indexed: 12/25/2022] Open
Abstract
Cerebral ischemia can occur at any stage in life, but clinical consequences greatly differ depending on the developmental stage of the affected brain structures. Timing of the lesion occurrence seems to be critical, as it strongly interferes with neuronal circuit development and determines the way spontaneous plasticity takes place. Translational stroke research requires the use of animal models as they represent a reliable tool to understand the pathogenic mechanisms underlying the generation, progression, and pathological consequences of a stroke. Moreover, in vivo experiments are instrumental to investigate new therapeutic strategies and the best temporal window of intervention. Differently from adults, very few models of the human developmental stroke have been characterized, and most of them have been established in rodents. The models currently used provide a better understanding of the molecular factors involved in the effects of ischemia; however, they still hold many limitations due to matching developmental stages across different species and the complexity of the human disorder that hardly can be described by segregated variables. In this review, we summarize the key factors contributing to neonatal brain vulnerability to ischemic strokes and we provide an overview of the advantages and limitations of the currently available models to recapitulate different aspects of the human developmental stroke.
Collapse
|
147
|
Cree BAC, Niu J, Hoi KK, Zhao C, Caganap SD, Henry RG, Dao DQ, Zollinger DR, Mei F, Shen YAA, Franklin RJM, Ullian EM, Xiao L, Chan JR, Fancy SPJ. Clemastine rescues myelination defects and promotes functional recovery in hypoxic brain injury. Brain 2019; 141:85-98. [PMID: 29244098 DOI: 10.1093/brain/awx312] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 10/14/2017] [Indexed: 11/14/2022] Open
Abstract
Hypoxia can injure brain white matter tracts, comprised of axons and myelinating oligodendrocytes, leading to cerebral palsy in neonates and delayed post-hypoxic leukoencephalopathy (DPHL) in adults. In these conditions, white matter injury can be followed by myelin regeneration, but myelination often fails and is a significant contributor to fixed demyelinated lesions, with ensuing permanent neurological injury. Non-myelinating oligodendrocyte precursor cells are often found in lesions in plentiful numbers, but fail to mature, suggesting oligodendrocyte precursor cell differentiation arrest as a critical contributor to failed myelination in hypoxia. We report a case of an adult patient who developed the rare condition DPHL and made a nearly complete recovery in the setting of treatment with clemastine, a widely available antihistamine that in preclinical models promotes oligodendrocyte precursor cell differentiation. This suggested possible therapeutic benefit in the more clinically prevalent hypoxic injury of newborns, and we demonstrate in murine neonatal hypoxic injury that clemastine dramatically promotes oligodendrocyte precursor cell differentiation, myelination, and improves functional recovery. We show that its effect in hypoxia is oligodendroglial specific via an effect on the M1 muscarinic receptor on oligodendrocyte precursor cells. We propose clemastine as a potential therapy for hypoxic brain injuries associated with white matter injury and oligodendrocyte precursor cell maturation arrest.
Collapse
Affiliation(s)
- Bruce A C Cree
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California at San Francisco, San Francisco, CA 94158, USA
| | - Jianqin Niu
- Department of Pediatrics, University of California at San Francisco, San Francisco, CA 94158, USA.,Department of Histology and Embryology, Collaborative Innovation Center for Brain Research, Third Military Medical University, Chongqing 400038, China
| | - Kimberly K Hoi
- Department of Pediatrics, University of California at San Francisco, San Francisco, CA 94158, USA
| | - Chao Zhao
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0AH, UK
| | - Scott D Caganap
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California at San Francisco, San Francisco, CA 94158, USA
| | - Roland G Henry
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California at San Francisco, San Francisco, CA 94158, USA
| | - Dang Q Dao
- Department of Ophthalmology, University of California at San Francisco, San Francisco, CA 94158, USA
| | - Daniel R Zollinger
- Department of Pediatrics, University of California at San Francisco, San Francisco, CA 94158, USA
| | - Feng Mei
- Department of Histology and Embryology, Collaborative Innovation Center for Brain Research, Third Military Medical University, Chongqing 400038, China
| | - Yun-An A Shen
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California at San Francisco, San Francisco, CA 94158, USA
| | - Robin J M Franklin
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0AH, UK
| | - Erik M Ullian
- Department of Ophthalmology, University of California at San Francisco, San Francisco, CA 94158, USA
| | - Lan Xiao
- Department of Histology and Embryology, Collaborative Innovation Center for Brain Research, Third Military Medical University, Chongqing 400038, China
| | - Jonah R Chan
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California at San Francisco, San Francisco, CA 94158, USA
| | - Stephen P J Fancy
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California at San Francisco, San Francisco, CA 94158, USA.,Department of Pediatrics, University of California at San Francisco, San Francisco, CA 94158, USA.,Division of Neonatology, University of California at San Francisco, San Francisco, CA 94158, USA.,Newborn Brain Research Institute, University of California at San Francisco, San Francisco, CA 94158, USA
| |
Collapse
|
148
|
Dingman AL, Rodgers KM, Dietz RM, Hickey SP, Frazier AP, Clevenger AC, Yonchek JC, Traystman RJ, Macklin WB, Herson PS. Oligodendrocyte Progenitor Cell Proliferation and Fate after White Matter Stroke in Juvenile and Adult Mice. Dev Neurosci 2019; 40:1-16. [PMID: 30861520 DOI: 10.1159/000496200] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 12/06/2018] [Indexed: 11/19/2022] Open
Abstract
The incidence of stroke in children is 2.4 per 100,000 person-years and results in long-term motor and cognitive disability. In ischemic stroke, white matter (WM) is frequently injured, but is relatively understudied compared to grey matter injury. Previous research suggests that the cellular response to WM ischemic injury is different at different ages. Little is known about whether WM repair mechanisms differ in children and adults. We utilized a model of focal ischemic WM injury to determine the oligodendrocyte (OL) response to focal WM ischemic injury in juvenile and adult mice. Methods: Juvenile (21-25 days of age) versus adult (2-3 months of age) mice underwent stereotaxic injection of the potent vasoconstrictor N5-(1-iminoethyhl)-L-ornithine (L-NIO) into the lateral corpus callosum (CC). Animals were sacrificed on postoperative day 3 (acute) or 21 (chronic). Cell birth-dating was performed acutely after WM stroke with 5-ethynyl-2-deoxyuridine (EdU) injected intraperitoneally. Immunohistochemistry was performed, as well as stereology, to measure injury volume. The acute oligodendrocyte progenitor cell (OPC) proliferation and the chronic OL cell fate were determined with immunohistochemistry. Compound action potentials were measured in the CC at acute and chronic time points. Results: Acutely WM injury volume was smaller in juveniles. There was significantly greater OPC proliferation in juvenile animals (acute) compared to adults, but newly born OLs did not survive and mature into myelinating cells at chronic time points. In addition, juveniles did not have improved histological or functional recovery when compared to adults. Protecting newly born OPCs is a potential therapeutic target in children with ischemic stroke.
Collapse
Affiliation(s)
- Andra L Dingman
- Division of Child Neurology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, USA,
| | - Krista M Rodgers
- Department of Anesthesiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Robert M Dietz
- Division of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Sean P Hickey
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Alexandra P Frazier
- Department of Anesthesiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Amy C Clevenger
- Division of Critical Care Medicine, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Joan C Yonchek
- Department of Anesthesiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Richard J Traystman
- Department of Anesthesiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Wendy B Macklin
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Paco S Herson
- Department of Anesthesiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| |
Collapse
|
149
|
Hoch-Kraft P, Trotter J, Gonsior C. Missing in Action: Dysfunctional RNA Metabolism in Oligodendroglial Cells as a Contributor to Neurodegenerative Diseases? Neurochem Res 2019; 45:566-579. [PMID: 30843138 DOI: 10.1007/s11064-019-02763-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 02/22/2019] [Accepted: 02/23/2019] [Indexed: 12/14/2022]
Abstract
The formation of myelin around axons by oligodendrocytes (OL) poses an enormous synthetic and energy challenge for the glial cell. Local translation of transcripts, including the mRNA for the essential myelin protein Myelin Basic Protein (MBP) at the site of myelin deposition has been recognised as an efficient mechanism to assure proper myelin sheath assembly. Oligodendroglial precursor cells (OPCs) form synapses with neurons and may localise many additional mRNAs in a similar fashion to synapses between neurons. In some diseases in which demyelination occurs, an abundance of OPCs is present but there is a failure to efficiently remyelinate and to synthesise MBP. This compromises axonal survival and function. OPCs are especially sensitive to cellular stress as occurring in neurodegenerative diseases, which can impinge on their ability to translate mRNAs into protein. Stress causes the build up of cytoplasmic stress granules (SG) in which many RNAs are sequestered and translationally stalled until the stress ceases. Chronic stress in particular could convert this initially protective reaction of the cell into damage, as persistence of SG may lead to pathological aggregate formation or long-term translation block of SG-associated RNAs. The recent recognition that many neurodegenerative diseases often exhibit an early white matter pathology with a proliferation of surviving OPCs, renders a study of the stress-associated processes in oligodendrocytes and OPCs especially relevant. Here, we discuss a potential dysfunction of RNA regulation in myelin diseases such as Multiple Sclerosis (MS) and Vanishing white matter disease (VWM) and potential contributions of OL dysfunction to neurodegenerative diseases such as Amyotrophic lateral sclerosis (ALS), Alzheimer's disease (AD) and Fragile X syndrome (FXS).
Collapse
Affiliation(s)
- Peter Hoch-Kraft
- Cellular Neurobiology, Institute for Developmental Biology and Neurobiology, Johannes Gutenberg-University of Mainz, Anselm-Franz-von-Bentzelweg 3, 55128, Mainz, Germany
| | - Jacqueline Trotter
- Cellular Neurobiology, Institute for Developmental Biology and Neurobiology, Johannes Gutenberg-University of Mainz, Anselm-Franz-von-Bentzelweg 3, 55128, Mainz, Germany
| | - Constantin Gonsior
- Cellular Neurobiology, Institute for Developmental Biology and Neurobiology, Johannes Gutenberg-University of Mainz, Anselm-Franz-von-Bentzelweg 3, 55128, Mainz, Germany.
| |
Collapse
|
150
|
Ma Q, Zhang L, Pearce WJ. MicroRNAs in brain development and cerebrovascular pathophysiology. Am J Physiol Cell Physiol 2019; 317:C3-C19. [PMID: 30840494 DOI: 10.1152/ajpcell.00022.2019] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
MicroRNAs (miRNAs) are a class of highly conserved non-coding RNAs with 21-25 nucleotides in length and play an important role in regulating gene expression at the posttranscriptional level via base-paring with complementary sequences of the 3'-untranslated region of the target gene mRNA, leading to either transcript degradation or translation inhibition. Brain-enriched miRNAs act as versatile regulators of brain development and function, including neural lineage and subtype determination, neurogenesis, synapse formation and plasticity, neural stem cell proliferation and differentiation, and responses to insults. Herein, we summarize the current knowledge regarding the role of miRNAs in brain development and cerebrovascular pathophysiology. We review recent progress of the miRNA-based mechanisms in neuronal and cerebrovascular development as well as their role in hypoxic-ischemic brain injury. These findings hold great promise, not just for deeper understanding of basic brain biology but also for building new therapeutic strategies for prevention and treatment of pathologies such as cerebral ischemia.
Collapse
Affiliation(s)
- Qingyi Ma
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine , Loma Linda, California
| | - Lubo Zhang
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine , Loma Linda, California
| | - William J Pearce
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine , Loma Linda, California
| |
Collapse
|