1
|
Efstathiou N, Koliakos G, Kantziou K, Kyriazis G, Slavakis A, Drossou V, Soubasi V. Kinetics of Circulating Progenitor Cells and Chemotactic Factors in Full-Term Neonates with Encephalopathy: Indications of Participation in the Endogenous Regenerative Process. Biomolecules 2025; 15:427. [PMID: 40149963 PMCID: PMC11940357 DOI: 10.3390/biom15030427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 02/24/2025] [Accepted: 03/12/2025] [Indexed: 03/29/2025] Open
Abstract
Preclinical studies have shown that progenitor cells (PCs) are mobilized toward injured tissues to ameliorate damage and contribute to regeneration. The exogenous therapeutic administration of PCs in children affected by neonatal encephalopathy (NE) is a promising, yet underreported, topic. In this prospective study, we investigated whether endogenous circulating progenitor cells (CPCs) are involved in intrinsic regeneration mechanisms following neonatal brain injury. Thirteen full-term infants with moderate/severe NE, eleven with perinatal stress, and twelve controls were enrolled. Blood samples were collected on days 1, 3, 9, 18, and 45, as well as at 8 and 24 months of life, and were analyzed with a focus on Endothelial Progenitor Cells, Haematopoietic Stem Cells, and Very Small Embryonic-Like Stem Cells, in addition to chemotactic factors (erythropoietin, IGF-1, and SDF-1). Correlations between CPCs, chemotactic factors, and brain injury were assessed using serum levels of brain injury biomarkers (S100B and neuron-specific enolase), brain MRIs, and Bayley III developmental scores. Increased brain injury biomarkers were followed by the upregulation of SDF-1 receptor and erythropoietin and, finally, by elevated CPCs. These findings suggest a potential endogenous regenerative effort, primarily observed in the moderate encephalopathy group, but this is suppressed in cases of severe brain injury. Mimicking and enhancing endogenous regeneration pathways in cases of failure-regarding cell type and timeframe-could provide a novel therapeutic model.
Collapse
Affiliation(s)
- Nikolaos Efstathiou
- 1st Neonatal Clinic and NICU, Hippokration General Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece
| | - Georgios Koliakos
- Biochemistry Department, Medical School, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Katerina Kantziou
- 1st Neonatal Clinic and NICU, Hippokration General Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece
| | - Georgios Kyriazis
- Immunology Department, Pulmonary Clinic, Papanikolaou General Hospital, Aristotle University of Thessaloniki, Exohi, 57010 Thessaloniki, Greece
| | - Aristeidis Slavakis
- Biochemistry Department, Hippokration General Hospital, 54642 Thessaloniki, Greece
| | - Vasiliki Drossou
- 1st Neonatal Clinic and NICU, Hippokration General Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece
| | - Vasiliki Soubasi
- 1st Neonatal Clinic and NICU, Hippokration General Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece
| |
Collapse
|
2
|
Wassink G, Cho KHT, Mathai S, Lear CA, Dean JM, Gunn AJ, Bennet L. White matter protection with insulin-like growth factor-1 after hypoxia-ischaemia in preterm foetal sheep. Brain Commun 2024; 6:fcae373. [PMID: 39507274 PMCID: PMC11539755 DOI: 10.1093/braincomms/fcae373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 09/02/2024] [Accepted: 10/22/2024] [Indexed: 11/08/2024] Open
Abstract
Perinatal hypoxia-ischaemia in extremely preterm infants is associated with long-term neurodevelopmental impairment, for which there is no specific treatment. Insulin-like growth factor-1 can reduce acute brain injury, but its effects on chronic white matter injury after hypoxia-ischaemia are unclear. Preterm-equivalent foetal sheep (0.6 gestation) received either sham-asphyxia or asphyxia induced by umbilical cord occlusion for 30 min, and recovered for either 3 or 35 days after asphyxia. The 35 day recovery groups received either an intracerebroventricular infusion of insulin-like growth factor-1 (1 µg/24 h) or vehicle, from 3 to 14 days after asphyxia. Asphyxia was associated with ventricular enlargement, and loss of frontal and parietal white matter area (P < 0.05 versus sham-asphyxia). This was associated with reduced area fraction of myelin basic protein and numbers of oligodendrocyte transcription factor 2 and mature, anti-adenomatous polyposis coli-positive oligodendrocytes in periventricular white matter (P < 0.05), with persistent inflammation and caspase-3 activation (P < 0.05). Four of eight foetuses developed cystic lesions in temporal white matter. Prolonged infusion with insulin-like growth factor-1 restored frontal white matter area, improved numbers of oligodendrocyte transcription factor 2-positive and mature, anti-adenomatous polyposis coli-positive oligodendrocytes, with reduced astrogliosis and microgliosis after 35 days recovery (P < 0.05 versus asphyxia). One of four foetuses developed temporal cystic lesions. Functionally, insulin-like growth factor-1-treated foetuses had faster recovery of EEG power, but not spectral edge. Encouragingly, these findings show that delayed, prolonged, insulin-like growth factor-1 treatment can improve functional maturation of periventricular white matter after severe asphyxia in the very immature brain, at least in part by suppressing chronic neural inflammation.
Collapse
Affiliation(s)
- Guido Wassink
- Department of Physiology, University of Auckland, Private Bag 92019, Auckland 1023, New Zealand
| | - Kenta H T Cho
- Department of Physiology, University of Auckland, Private Bag 92019, Auckland 1023, New Zealand
| | - Sam Mathai
- Department of Physiology, University of Auckland, Private Bag 92019, Auckland 1023, New Zealand
| | - Christopher A Lear
- Department of Physiology, University of Auckland, Private Bag 92019, Auckland 1023, New Zealand
| | - Justin M Dean
- Department of Physiology, University of Auckland, Private Bag 92019, Auckland 1023, New Zealand
| | - Alistair J Gunn
- Department of Physiology, University of Auckland, Private Bag 92019, Auckland 1023, New Zealand
| | - Laura Bennet
- Department of Physiology, University of Auckland, Private Bag 92019, Auckland 1023, New Zealand
| |
Collapse
|
3
|
Gunn AJ, Davidson JO. Stay cool and keep moving forwards. Pediatr Res 2024:10.1038/s41390-024-03546-0. [PMID: 39242940 DOI: 10.1038/s41390-024-03546-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 08/20/2024] [Accepted: 08/24/2024] [Indexed: 09/09/2024]
Affiliation(s)
- Alistair J Gunn
- Department of Physiology, The University of Auckland, Auckland, New Zealand.
| | - Joanne O Davidson
- Department of Physiology, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
4
|
Fleiss B, Gressens P. Role of Microglial Modulation in Therapies for Perinatal Brain Injuries Leading to Neurodevelopmental Disorders. ADVANCES IN NEUROBIOLOGY 2024; 37:591-606. [PMID: 39207715 DOI: 10.1007/978-3-031-55529-9_33] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Neurodevelopmental disorders (NDDs) encompass various conditions stemming from changes during brain development, typically diagnosed early in life. Examples include autism spectrum disorder, intellectual disability, cerebral palsy, seizures, dyslexia, and attention deficit hyperactivity disorder. Many NDDs are linked to perinatal events like infections, oxygen disturbances, or insults in combination. This chapter outlines the causes and effects of perinatal brain injury as they relate to microglia, along with efforts to prevent or treat such damage. We primarily discuss therapies targeting microglia modulation, focusing on those either clinically used or in advanced development, often tested in large animal models such as sheep, non-human primates, and piglets-standard translational models in perinatal medicine. Additionally, it touches on experimental studies showcasing advancements in the field.
Collapse
Affiliation(s)
- Bobbi Fleiss
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
- Université de Paris, NeuroDiderot, Inserm, Paris, France
| | - Pierre Gressens
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia.
- Université de Paris, NeuroDiderot, Inserm, Paris, France.
| |
Collapse
|
5
|
Du H, Xia J, Huang L, Zheng L, Gu W, Yi F. Relationship between insulin-like growth factor-1 and cerebral small vessel disease and its mechanisms: advances in the field. Front Aging Neurosci 2023; 15:1190869. [PMID: 37358957 PMCID: PMC10285072 DOI: 10.3389/fnagi.2023.1190869] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 05/23/2023] [Indexed: 06/28/2023] Open
Abstract
Insulin-like growth factor-1 (IGF-1) is an active polypeptide protein that closely resembles the structural sequence of insulin and is involved in a variety of metabolic processes in the body. Decreased IGF-1 circulation levels are associated with an increased risk of stroke and a poorer prognosis, but the relationship with cerebral small vessel disease (cSVD) is unclear. Some studies found that the level of IGF-1 in patients with cSVD was significantly reduced, but the clinical significance and underlying mechanisms are unknown. This article reviews the correlation between IGF-1 and cerebrovascular disease and explores the potential relationship and mechanism between IGF-1 and cSVD.
Collapse
Affiliation(s)
- Hao Du
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jian Xia
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Clinical Research Center for Cerebrovascular Disease of Hunan Province, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lei Huang
- Department of Rehabilitation, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, China
| | - Lan Zheng
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wenping Gu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Clinical Research Center for Cerebrovascular Disease of Hunan Province, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Fang Yi
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Geriatric Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
6
|
Gaire BP. Microglia as the Critical Regulators of Neuroprotection and Functional Recovery in Cerebral Ischemia. Cell Mol Neurobiol 2022; 42:2505-2525. [PMID: 34460037 PMCID: PMC11421653 DOI: 10.1007/s10571-021-01145-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 08/25/2021] [Indexed: 12/13/2022]
Abstract
Microglial activation is considered as the critical pathogenic event in diverse central nervous system disorders including cerebral ischemia. Proinflammatory responses of activated microglia have been well reported in the ischemic brain and neuroinflammatory responses of activated microglia have been believed to be the potential therapeutic strategy. However, despite having proinflammatory roles, microglia can have significant anti-inflammatory roles and they are associated with the production of growth factors which are responsible for neuroprotection and recovery after ischemic injury. Microglia can directly promote neuroprotection by preventing ischemic infarct expansion and promoting functional outcomes. Indirectly, microglia are involved in promoting anti-inflammatory responses, neurogenesis, and angiogenesis in the ischemic brain which are crucial pathophysiological events for ischemic recovery. In fact, anti-inflammatory cytokines and growth factors produced by microglia can promote neuroprotection and attenuate neurobehavioral deficits. In addition, microglia regulate phagocytosis, axonal regeneration, blood-brain barrier protection, white matter integrity, and synaptic remodeling, which are essential for ischemic recovery. Microglia can also regulate crosstalk with neurons and other cell types to promote neuroprotection and ischemic recovery. This review mainly focuses on the roles of microglia in neuroprotection and recovery following ischemic injury. Furthermore, this review also sheds the light on the therapeutic potential of microglia in stroke patients.
Collapse
Affiliation(s)
- Bhakta Prasad Gaire
- Department of Neurology and Anesthesiology, Shock Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
| |
Collapse
|
7
|
Motavaf M, Piao X. Oligodendrocyte Development and Implication in Perinatal White Matter Injury. Front Cell Neurosci 2021; 15:764486. [PMID: 34803612 PMCID: PMC8599582 DOI: 10.3389/fncel.2021.764486] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 10/11/2021] [Indexed: 12/12/2022] Open
Abstract
Perinatal white matter injury (WMI) is the most common brain injury in premature infants and can lead to life-long neurological deficits such as cerebral palsy. Preterm birth is typically accompanied by inflammation and hypoxic-ischemic events. Such perinatal insults negatively impact maturation of oligodendrocytes (OLs) and cause myelination failure. At present, no treatment options are clinically available to prevent or cure WMI. Given that arrested OL maturation plays a central role in the etiology of perinatal WMI, an increased interest has emerged regarding the functional restoration of these cells as potential therapeutic strategy. Cell transplantation and promoting endogenous oligodendrocyte function are two potential options to address this major unmet need. In this review, we highlight the underlying pathophysiology of WMI with a specific focus on OL biology and their implication for the development of new therapeutic targets.
Collapse
Affiliation(s)
- Mahsa Motavaf
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Xianhua Piao
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, United States.,Newborn Brain Research Institute, University of California, San Francisco, San Francisco, CA, United States.,Weill Institute for Neuroscience, University of California, San Francisco, San Francisco, CA, United States.,Division of Neonatology, Department of Pediatrics, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
8
|
Blum K, Gold MS, Llanos-Gomez L, Jalali R, Thanos PK, Bowirrat A, Downs WB, Bagchi D, Braverman ER, Baron D, Roy AK, Badgaiyan RD. Hypothesizing Nutrigenomic-Based Precision Anti-Obesity Treatment and Prophylaxis: Should We Be Targeting Sarcopenia Induced Brain Dysfunction? INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18189774. [PMID: 34574696 PMCID: PMC8470221 DOI: 10.3390/ijerph18189774] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/27/2021] [Accepted: 08/28/2021] [Indexed: 12/15/2022]
Abstract
Background: The United States Centers for Disease Control and Prevention (CDC) estimates a total obesity rate of 30% for 12 states and a 20% obesity rate nationwide. The obesity epidemic continues to increase in spite of preventative measures undertaken worldwide. Pharmacological treatments promise to reduce total fat mass. However, medications may have significant side effects and can be potentially fatal. Data Retrieval: This brief review, based on a PUBMED search of the key terms "Obesity" and" Sarcopenia," will present evidence to corroborate the existence of Reward Deficiency Syndrome (RDS) in obesity and the involvement of catecholaminergic pathways in substance seeking behavior, particularly as it relates to carbohydrates cravings. Expert Opinion: The genetic basis and future genetic testing of children for risk of aberrant generalized craving behavior are considered a prevention method. Here we present evidence supporting the use of precursor amino acid therapy and modulation of enkephalinase, MOA, and COMT inhibition in key brain regions. Such treatments manifest in improved levels of dopamine/norepinephrine, GABA, serotonin, and enkephalins. We also present evidence substantiating insulin sensitivity enhancement via Chromium salts, which affect dopamine neuronal synthesis regulation. We believe our unique combination of natural ingredients will influence many pathways leading to the promotion of well-being and normal healthy metabolic functioning. Sarcopenia has been shown to reduce angiogenesis and possible cerebral blood flow. Exercise seems to provide a significant benefit to overcome this obesity-promoting loss of muscle density. Conclusion: Utilization of proposed nutrigenomic formulae based on coupling genetic obesity risk testing promotes generalized anti-craving of carbohydrates and can inhibit carbohydrate bingeing, inducing significant healthy fat loss and relapse prevention.
Collapse
Affiliation(s)
- Kenneth Blum
- Center for Psychiatry, Medicine & Primary Care (Office of the Provost), Division of Addiction Research & Education, Western University Health Science, Pomona, CA 91766, USA;
- Institute of Psychology, ELTE Eötvös Loránd University, Kazinczy u. 23-27, 1075 Budapest, Hungary
- Division of Nutrigenomics, Genomic Testing Center Geneus Health, San Antonio, TX 78249, USA
- Department of Psychiatry, University of Vermont, Burlington, VT 05401, USA
- Department of Psychiatry, Wright University Boonshoff School of Medicine, Dayton, OH 45377, USA
- Division of Precision Nutrition, Victory Nutrition International, Bonita Springs, FL 34135, USA; (W.B.D.); (D.B.)
- The Kenneth Blum Behavioral & Neurogenetic Institute, Division of Ivitalize Inc., Austin, TX 78701, USA; (L.L.-G.); (R.J.)
- Division of Clinical Neurology, Path Foundation NY, New York, NY 10010, USA;
- Correspondence:
| | - Mark S. Gold
- Department of Psychiatry, School of Medicine, Tulane University, New Orleans, LA 70118, USA; (M.S.G.); (A.K.R.III)
| | - Luis Llanos-Gomez
- The Kenneth Blum Behavioral & Neurogenetic Institute, Division of Ivitalize Inc., Austin, TX 78701, USA; (L.L.-G.); (R.J.)
| | - Rehan Jalali
- The Kenneth Blum Behavioral & Neurogenetic Institute, Division of Ivitalize Inc., Austin, TX 78701, USA; (L.L.-G.); (R.J.)
| | - Panayotis K. Thanos
- Department of Psychology & Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions (BNNLA), Research Institute on Addictions, University at Buffalo, Buffalo, NY 14260, USA;
| | - Abdalla Bowirrat
- Department of Molecular Biology, Adelson School of Medicine, Ariel University, Ariel 40700, Israel;
| | - William B. Downs
- Division of Precision Nutrition, Victory Nutrition International, Bonita Springs, FL 34135, USA; (W.B.D.); (D.B.)
| | - Debasis Bagchi
- Division of Precision Nutrition, Victory Nutrition International, Bonita Springs, FL 34135, USA; (W.B.D.); (D.B.)
- Department of Pharmaceutical Sciences, College of Pharmacy & Health Sciences, Texas Southern University, Houston, TX 77004, USA
| | - Eric R. Braverman
- Division of Clinical Neurology, Path Foundation NY, New York, NY 10010, USA;
| | - David Baron
- Center for Psychiatry, Medicine & Primary Care (Office of the Provost), Division of Addiction Research & Education, Western University Health Science, Pomona, CA 91766, USA;
| | - Alphonso Kenison Roy
- Department of Psychiatry, School of Medicine, Tulane University, New Orleans, LA 70118, USA; (M.S.G.); (A.K.R.III)
| | - Rajendra D. Badgaiyan
- Department of Psychiatry, South Texas Veteran Health Care System, Audie L. Murphy Memorial VA Hospital, Long School of Medicine, University of Texas Medical Center, San Antonio, TX 78249, USA;
| |
Collapse
|
9
|
Positive Association Between Serum Insulin-Like Growth Factor-1 and Cognition in Patients with Cerebral Small Vessel Disease. J Stroke Cerebrovasc Dis 2021; 30:105790. [PMID: 33878547 DOI: 10.1016/j.jstrokecerebrovasdis.2021.105790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 03/14/2021] [Accepted: 03/24/2021] [Indexed: 11/20/2022] Open
Abstract
Cognitive impairment is one of the main complications of cerebral small vessel disease (CSVD). Serum insulin-like growth factor-1 (IGF-1) might serve as a marker for the risk of cognitive decline in patients with CSVD. We investigated the association of IGF-1 with the development of cognitive impairment in patients with CSVD. We included 216 patients with CVSD (mean age, 67.57 ± 8.53 years; 31.9% female). We compared 117 (54.2%) patients who developed cognitive impairment with 99 (45.8%) patients without cognitive impairment. Patients who developed cognitive impairment had significantly lower levels of IGF-I (p < 0 .001), suggesting that altered IGF-1 signaling may be a risk factor for cognitive decline in patients with CSVD.
Collapse
|
10
|
Breton JM, Long KLP, Barraza MK, Perloff OS, Kaufer D. Hormonal Regulation of Oligodendrogenesis II: Implications for Myelin Repair. Biomolecules 2021; 11:290. [PMID: 33669242 PMCID: PMC7919830 DOI: 10.3390/biom11020290] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 02/10/2021] [Accepted: 02/13/2021] [Indexed: 02/07/2023] Open
Abstract
Alterations in myelin, the protective and insulating sheath surrounding axons, affect brain function, as is evident in demyelinating diseases where the loss of myelin leads to cognitive and motor dysfunction. Recent evidence suggests that changes in myelination, including both hyper- and hypo-myelination, may also play a role in numerous neurological and psychiatric diseases. Protecting myelin and promoting remyelination is thus crucial for a wide range of disorders. Oligodendrocytes (OLs) are the cells that generate myelin, and oligodendrogenesis, the creation of new OLs, continues throughout life and is necessary for myelin plasticity and remyelination. Understanding the regulation of oligodendrogenesis and myelin plasticity within disease contexts is, therefore, critical for the development of novel therapeutic targets. In our companion manuscript, we review literature demonstrating that multiple hormone classes are involved in the regulation of oligodendrogenesis under physiological conditions. The majority of hormones enhance oligodendrogenesis, increasing oligodendrocyte precursor cell differentiation and inducing maturation and myelin production in OLs. Thus, hormonal treatments present a promising route to promote remyelination. Here, we review the literature on hormonal regulation of oligodendrogenesis within the context of disorders. We focus on steroid hormones, including glucocorticoids and sex hormones, peptide hormones such as insulin-like growth factor 1, and thyroid hormones. For each hormone, we describe whether they aid in OL survival, differentiation, or remyelination, and we discuss their mechanisms of action, if known. Several of these hormones have yielded promising results in both animal models and in human conditions; however, a better understanding of hormonal effects, interactions, and their mechanisms will ultimately lead to more targeted therapeutics for myelin repair.
Collapse
Affiliation(s)
- Jocelyn M Breton
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA 94720, USA
| | - Kimberly L P Long
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA 94720, USA
| | - Matthew K Barraza
- Molecular and Cellular Biology, University of California Berkeley, Berkeley, CA 94720, USA
| | - Olga S Perloff
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Daniela Kaufer
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA 94720, USA
- Integrative Biology, University of California Berkeley, Berkeley, CA 94720, USA
- Canadian Institute for Advanced Research, Toronto, ON M5G1M1, Canada
| |
Collapse
|
11
|
Vaes JEG, Kosmeijer CM, Kaal M, van Vliet R, Brandt MJV, Benders MJNL, Nijboer CH. Regenerative Therapies to Restore Interneuron Disturbances in Experimental Models of Encephalopathy of Prematurity. Int J Mol Sci 2020; 22:ijms22010211. [PMID: 33379239 PMCID: PMC7795049 DOI: 10.3390/ijms22010211] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 12/22/2020] [Accepted: 12/23/2020] [Indexed: 12/22/2022] Open
Abstract
Encephalopathy of Prematurity (EoP) is a major cause of morbidity in (extreme) preterm neonates. Though the majority of EoP research has focused on failure of oligodendrocyte maturation as an underlying pathophysiological mechanism, recent pioneer work has identified developmental disturbances in inhibitory interneurons to contribute to EoP. Here we investigated interneuron abnormalities in two experimental models of EoP and explored the potential of two promising treatment strategies, namely intranasal mesenchymal stem cells (MSCs) or insulin-like growth factor I (IGF1), to restore interneuron development. In rats, fetal inflammation and postnatal hypoxia led to a transient increase in total cortical interneuron numbers, with a layer-specific deficit in parvalbumin (PV)+ interneurons. Additionally, a transient excess of total cortical cell density was observed, including excitatory neuron numbers. In the hippocampal cornu ammonis (CA) 1 region, long-term deficits in total interneuron numbers and PV+ subtype were observed. In mice subjected to postnatal hypoxia/ischemia and systemic inflammation, total numbers of cortical interneurons remained unaffected; however, subtype analysis revealed a global, transient reduction in PV+ cells and a long-lasting layer-specific increase in vasoactive intestinal polypeptide (VIP)+ cells. In the dentate gyrus, a long-lasting deficit of somatostatin (SST)+ cells was observed. Both intranasal MSC and IGF1 therapy restored the majority of interneuron abnormalities in EoP mice. In line with the histological findings, EoP mice displayed impaired social behavior, which was partly restored by the therapies. In conclusion, induction of experimental EoP is associated with model-specific disturbances in interneuron development. In addition, intranasal MSCs and IGF1 are promising therapeutic strategies to aid interneuron development after EoP.
Collapse
Affiliation(s)
- Josine E. G. Vaes
- Department for Developmental Origins of Disease, University Medical Center Utrecht Brain Center and Wilhelmina Children’s Hospital, Utrecht University, 3584 Utrecht, The Netherlands; (J.E.G.V.); (C.M.K.); (M.K.); (R.v.V.); (M.J.V.B.)
- Department of Neonatology, University Medical Center Utrecht Brain Center and Wilhelmina Children’s Hospital, Utrecht University, 3584 Utrecht, The Netherlands;
| | - Chantal M. Kosmeijer
- Department for Developmental Origins of Disease, University Medical Center Utrecht Brain Center and Wilhelmina Children’s Hospital, Utrecht University, 3584 Utrecht, The Netherlands; (J.E.G.V.); (C.M.K.); (M.K.); (R.v.V.); (M.J.V.B.)
| | - Marthe Kaal
- Department for Developmental Origins of Disease, University Medical Center Utrecht Brain Center and Wilhelmina Children’s Hospital, Utrecht University, 3584 Utrecht, The Netherlands; (J.E.G.V.); (C.M.K.); (M.K.); (R.v.V.); (M.J.V.B.)
| | - Rik van Vliet
- Department for Developmental Origins of Disease, University Medical Center Utrecht Brain Center and Wilhelmina Children’s Hospital, Utrecht University, 3584 Utrecht, The Netherlands; (J.E.G.V.); (C.M.K.); (M.K.); (R.v.V.); (M.J.V.B.)
| | - Myrna J. V. Brandt
- Department for Developmental Origins of Disease, University Medical Center Utrecht Brain Center and Wilhelmina Children’s Hospital, Utrecht University, 3584 Utrecht, The Netherlands; (J.E.G.V.); (C.M.K.); (M.K.); (R.v.V.); (M.J.V.B.)
| | - Manon J. N. L. Benders
- Department of Neonatology, University Medical Center Utrecht Brain Center and Wilhelmina Children’s Hospital, Utrecht University, 3584 Utrecht, The Netherlands;
| | - Cora H. Nijboer
- Department for Developmental Origins of Disease, University Medical Center Utrecht Brain Center and Wilhelmina Children’s Hospital, Utrecht University, 3584 Utrecht, The Netherlands; (J.E.G.V.); (C.M.K.); (M.K.); (R.v.V.); (M.J.V.B.)
- Correspondence: ; Tel.: +31-88-755-4360
| |
Collapse
|
12
|
Vaes JEG, Brandt MJV, Wanders N, Benders MJNL, de Theije CGM, Gressens P, Nijboer CH. The impact of trophic and immunomodulatory factors on oligodendrocyte maturation: Potential treatments for encephalopathy of prematurity. Glia 2020; 69:1311-1340. [PMID: 33595855 PMCID: PMC8246971 DOI: 10.1002/glia.23939] [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: 09/13/2020] [Revised: 11/11/2020] [Accepted: 11/12/2020] [Indexed: 12/11/2022]
Abstract
Encephalopathy of prematurity (EoP) is a major cause of morbidity in preterm neonates, causing neurodevelopmental adversities that can lead to lifelong impairments. Preterm birth-related insults, such as cerebral oxygen fluctuations and perinatal inflammation, are believed to negatively impact brain development, leading to a range of brain abnormalities. Diffuse white matter injury is a major hallmark of EoP and characterized by widespread hypomyelination, the result of disturbances in oligodendrocyte lineage development. At present, there are no treatment options available, despite the enormous burden of EoP on patients, their families, and society. Over the years, research in the field of neonatal brain injury and other white matter pathologies has led to the identification of several promising trophic factors and cytokines that contribute to the survival and maturation of oligodendrocytes, and/or dampening neuroinflammation. In this review, we discuss the current literature on selected factors and their therapeutic potential to combat EoP, covering a wide range of in vitro, preclinical and clinical studies. Furthermore, we offer a future perspective on the translatability of these factors into clinical practice.
Collapse
Affiliation(s)
- Josine E G Vaes
- Department for Developmental Origins of Disease, University Medical Center Utrecht Brain Center and Wilhelmina Children's Hospital, Utrecht University, Utrecht, The Netherlands.,Department of Neonatology, University Medical Center Utrecht Brain Center and Wilhelmina Children's Hospital, Utrecht University, Utrecht, The Netherlands
| | - Myrna J V Brandt
- Department for Developmental Origins of Disease, University Medical Center Utrecht Brain Center and Wilhelmina Children's Hospital, Utrecht University, Utrecht, The Netherlands
| | - Nikki Wanders
- Department for Developmental Origins of Disease, University Medical Center Utrecht Brain Center and Wilhelmina Children's Hospital, Utrecht University, Utrecht, The Netherlands
| | - Manon J N L Benders
- Department of Neonatology, University Medical Center Utrecht Brain Center and Wilhelmina Children's Hospital, Utrecht University, Utrecht, The Netherlands
| | - Caroline G M de Theije
- Department for Developmental Origins of Disease, University Medical Center Utrecht Brain Center and Wilhelmina Children's Hospital, Utrecht University, Utrecht, The Netherlands
| | | | - Cora H Nijboer
- Department for Developmental Origins of Disease, University Medical Center Utrecht Brain Center and Wilhelmina Children's Hospital, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
13
|
Zhou KQ, Davidson JO, Bennet L, Gunn AJ. Combination treatments with therapeutic hypothermia for hypoxic-ischemic neuroprotection. Dev Med Child Neurol 2020; 62:1131-1137. [PMID: 32614467 DOI: 10.1111/dmcn.14610] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/15/2020] [Indexed: 12/12/2022]
Abstract
Therapeutic hypothermia is now proven to reduce death or disability in term and near-term born infants with moderate to severe hypoxic-ischemic encephalopathy. Nevertheless, many infants still survive with disability, despite treatment with hypothermia. Recent preclinical and clinical studies suggest that current protocols for therapeutic hypothermia are near-optimal. The obvious strategy, in addition to improving early initiation of therapeutic hypothermia after birth, is to combine hypothermia with other neuroprotective agents. We review evidence that the mechanisms of action of many promising agents overlap with the anti-excitotoxic, anti-apoptotic, and anti-inflammatory mechanisms of hypothermia, leading to a lack of benefit from combination treatment. Moreover, even apparently beneficial combinations have failed to translate in clinical trials. These considerations highlight the need for preclinical studies to test clinically realistic protocols of timing and duration of treatment, before committing to large randomized controlled trials.
Collapse
Affiliation(s)
- Kelly Q Zhou
- Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Joanne O Davidson
- Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Laura Bennet
- Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Alistair J Gunn
- Department of Physiology, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
14
|
Youssef MI, Zhou Y, Eissa IH, Wang Y, Zhang J, Jiang L, Hu W, Qi J, Chen Z. Tetradecyl 2,3-dihydroxybenzoate alleviates oligodendrocyte damage following chronic cerebral hypoperfusion through IGF-1 receptor. Neurochem Int 2020; 138:104749. [PMID: 32387468 DOI: 10.1016/j.neuint.2020.104749] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 04/08/2020] [Accepted: 04/23/2020] [Indexed: 12/21/2022]
Abstract
Currently, there is no effective therapy for chronic cerebral hypoperfusion-induced subcortical ischemic vascular dementia (SIVD), which displays cognitive deficits and progressive white matter damage. Tetradecyl 2,3-dihydroxybenzoate (ABG-001) is a lead compound derived from gentisides with neuritogenic activity. In this report, we intended to investigate the effect of ABG-001 on the SIVD experimental model through right unilateral common carotid arteries occlusion (rUCCAO) in mice. We found that ABG-001 remarkably alleviated white matter damage and cognitive deficits after cerebral hypoperfusion induced by rUCCAO. The protection of ABG-001 on the white matter was related to an amelioration of the oligodendrocyte apoptosis and demyelination rather than promoting remyelination. Molecular docking study showed that ABG-001 possesses a high affinity for insulin-like growth factor-1 receptor (IGF-1R), but not for tropomyosin receptor kinase A (TrkA). The protection of ABG-001 against oligodendrocyte damage was abrogated by IGF-1R antagonist or knockdown of IGF-1R through shRNA, but not TrkA antagonist. Moreover, ABG-001 did not induce hematological, renal or hepatic toxicity after chronic treatment. The present study indicates that ABG-001 protects oligodendrocytes through IGF-1R to relieve demyelination following chronic cerebral hypoperfusion, which could be represented as an encouraging treatment for SIVD.
Collapse
Affiliation(s)
- Mahmoud I Youssef
- Institute of Pharmacology & Toxicology, NHC and CAMS Key Laboratory of Medical Neurobiology, College of Pharmaceutical Sciences, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310058, PR China
| | - Yiting Zhou
- Institute of Pharmacology & Toxicology, NHC and CAMS Key Laboratory of Medical Neurobiology, College of Pharmaceutical Sciences, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310058, PR China; Department of Pharmacy, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, East Qingchun Road 3, Hangzhou, Zhejiang, 310016, PR China
| | - Ibrahim H Eissa
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, 11884, Egypt
| | - Yanhui Wang
- Institute of Pharmacology & Toxicology, NHC and CAMS Key Laboratory of Medical Neurobiology, College of Pharmaceutical Sciences, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310058, PR China
| | - Jing Zhang
- Department of Pharmacy, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, East Qingchun Road 3, Hangzhou, Zhejiang, 310016, PR China
| | - Lei Jiang
- Institute of Pharmacology & Toxicology, NHC and CAMS Key Laboratory of Medical Neurobiology, College of Pharmaceutical Sciences, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310058, PR China
| | - Weiwei Hu
- Institute of Pharmacology & Toxicology, NHC and CAMS Key Laboratory of Medical Neurobiology, College of Pharmaceutical Sciences, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310058, PR China.
| | - Jianhua Qi
- Institute of Pharmacology & Toxicology, NHC and CAMS Key Laboratory of Medical Neurobiology, College of Pharmaceutical Sciences, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310058, PR China
| | - Zhong Chen
- Institute of Pharmacology & Toxicology, NHC and CAMS Key Laboratory of Medical Neurobiology, College of Pharmaceutical Sciences, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310058, PR China.
| |
Collapse
|
15
|
Serhan A, Boddeke E, Kooijman R. Insulin-Like Growth Factor-1 Is Neuroprotective in Aged Rats With Ischemic Stroke. Front Aging Neurosci 2019; 11:349. [PMID: 31920629 PMCID: PMC6918863 DOI: 10.3389/fnagi.2019.00349] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 11/29/2019] [Indexed: 01/01/2023] Open
Abstract
Post-stroke systemic injections of insulin-like growth factor-1 (IGF-1) exert neuroprotective effects in rats. In the current study, we aimed to test the efficacy of IGF-1 neuroprotection in aged rats (24–25 months old) and to compare the results with adult rats (6–7 months old). Furthermore, we addressed putative differences in microglial responses to IGF-1 in adult and aged rats. Rats were subjected to ischemic stroke while they were conscious by infusing endothelin-1 (Et-1) through a guide cannula that was implemented in the vicinity of the middle cerebral artery (MCA). Rats were given subcutaneous injections of IGF-1 (1 mg/kg) at 30 min and 120 min after the insult. Post-stroke IGF-1 treatment reduced the infarct size by 34% and 38% in aged and adult rats, respectively. The IGF-1 treated adult rats also showed significant improvement in sensorimotor function following stroke, while this function was not significantly affected in aged rats. Furthermore, aged rats displayed exaggerated activation of microglia in the ischemic hemisphere. Significant reduction of microglial activation by IGF-1 was only detected at specific regions in the ipsilateral hemisphere of adult rats. We show that IGF-1 reduced infarct size in aged rats with an ischemic stroke. It remains to be established, however, whether the age-related changes in microglial function affect the improvement in behavioral outcomes.
Collapse
Affiliation(s)
- Ahmad Serhan
- Department of Experimental Pharmacology, Center for Neurosciences (C4N), Vrije Universiteit Brussel, Brussels, Belgium.,Department of Neuroscience, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Erik Boddeke
- Department of Neuroscience, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Ron Kooijman
- Department of Experimental Pharmacology, Center for Neurosciences (C4N), Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
16
|
Developing Trojan horses to induce, diagnose and suppress Alzheimer’s pathology. Pharmacol Res 2019; 149:104471. [DOI: 10.1016/j.phrs.2019.104471] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 09/17/2019] [Accepted: 09/30/2019] [Indexed: 01/05/2023]
|
17
|
Morrison JL, Berry MJ, Botting KJ, Darby JRT, Frasch MG, Gatford KL, Giussani DA, Gray CL, Harding R, Herrera EA, Kemp MW, Lock MC, McMillen IC, Moss TJ, Musk GC, Oliver MH, Regnault TRH, Roberts CT, Soo JY, Tellam RL. Improving pregnancy outcomes in humans through studies in sheep. Am J Physiol Regul Integr Comp Physiol 2018; 315:R1123-R1153. [PMID: 30325659 DOI: 10.1152/ajpregu.00391.2017] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Experimental studies that are relevant to human pregnancy rely on the selection of appropriate animal models as an important element in experimental design. Consideration of the strengths and weaknesses of any animal model of human disease is fundamental to effective and meaningful translation of preclinical research. Studies in sheep have made significant contributions to our understanding of the normal and abnormal development of the fetus. As a model of human pregnancy, studies in sheep have enabled scientists and clinicians to answer questions about the etiology and treatment of poor maternal, placental, and fetal health and to provide an evidence base for translation of interventions to the clinic. The aim of this review is to highlight the advances in perinatal human medicine that have been achieved following translation of research using the pregnant sheep and fetus.
Collapse
Affiliation(s)
- Janna L Morrison
- Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Mary J Berry
- Department of Paediatrics and Child Health, University of Otago , Wellington , New Zealand
| | - Kimberley J Botting
- Department of Physiology, Development, and Neuroscience, University of Cambridge , Cambridge , United Kingdom
| | - Jack R T Darby
- Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Martin G Frasch
- Department of Obstetrics and Gynecology, University of Washington , Seattle, Washington
| | - Kathryn L Gatford
- Robinson Research Institute and Adelaide Medical School, University of Adelaide , Adelaide, South Australia , Australia
| | - Dino A Giussani
- Department of Physiology, Development, and Neuroscience, University of Cambridge , Cambridge , United Kingdom
| | - Clint L Gray
- Department of Paediatrics and Child Health, University of Otago , Wellington , New Zealand
| | - Richard Harding
- Department of Anatomy and Developmental Biology, Monash University , Clayton, Victoria , Australia
| | - Emilio A Herrera
- Pathophysiology Program, Biomedical Sciences Institute (ICBM), Faculty of Medicine, University of Chile , Santiago , Chile
| | - Matthew W Kemp
- Division of Obstetrics and Gynecology, University of Western Australia , Perth, Western Australia , Australia
| | - Mitchell C Lock
- Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - I Caroline McMillen
- Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Timothy J Moss
- The Ritchie Centre, Hudson Institute of Medical Research, Department of Obstetrics and Gynaecology, Monash University , Clayton, Victoria , Australia
| | - Gabrielle C Musk
- Animal Care Services, University of Western Australia , Perth, Western Australia , Australia
| | - Mark H Oliver
- Liggins Institute, University of Auckland , Auckland , New Zealand
| | - Timothy R H Regnault
- Department of Obstetrics and Gynecology and Department of Physiology and Pharmacology, Western University, and Children's Health Research Institute , London, Ontario , Canada
| | - Claire T Roberts
- Robinson Research Institute and Adelaide Medical School, University of Adelaide , Adelaide, South Australia , Australia
| | - Jia Yin Soo
- Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Ross L Tellam
- Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia
| |
Collapse
|
18
|
Carlson SW, Saatman KE. Central Infusion of Insulin-Like Growth Factor-1 Increases Hippocampal Neurogenesis and Improves Neurobehavioral Function after Traumatic Brain Injury. J Neurotrauma 2018; 35:1467-1480. [PMID: 29455576 PMCID: PMC5998830 DOI: 10.1089/neu.2017.5374] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Traumatic brain injury (TBI) produces neuronal dysfunction and cellular loss that can culminate in lasting impairments in cognitive and motor abilities. Therapeutic agents that promote repair and replenish neurons post-TBI hold promise in improving recovery of function. Insulin-like growth factor-1 (IGF-1) is a neurotrophic factor capable of mediating neuroprotective and neuroplasticity mechanisms. Targeted overexpression of IGF-1 enhances the generation of hippocampal newborn neurons in brain-injured mice; however, the translational neurogenic potential of exogenously administered IGF-1 post-TBI remains unknown. In a mouse model of controlled cortical impact, continuous intracerebroventricular infusion of recombinant human IGF-1 (hIGF) for 7 days, beginning 15 min post-injury, resulted in a dose-dependent increase in the number of immature neurons in the hippocampus. Infusion of 10 μg/day of IGF-1 produced detectable levels of hIGF-1 in the cortex and hippocampus and a concomitant increase in protein kinase B activation in the hippocampus. Both motor function and cognition were improved over 7 days post-injury in IGF-1-treated cohorts. Vehicle-treated brain-injured mice showed reduced hippocampal immature neuron density relative to sham controls at 7 days post-injury. In contrast, the density of hippocampal immature neurons in brain-injured mice receiving acute onset IGF-1 infusion was significantly higher than in injured mice receiving vehicle and equivalent to that in sham-injured control mice. Importantly, the neurogenic effect of IGF-1 was maintained with as much as a 6-h delay in the initiation of infusion. These data suggest that central infusion of IGF-1 enhances the generation of immature neurons in the hippocampus, with a therapeutic window of at least 6 h post-injury, and promotes neurobehavioral recovery post-TBI.
Collapse
Affiliation(s)
- Shaun W. Carlson
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky
| | - Kathryn E. Saatman
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
19
|
Wassink G, Davidson JO, Lear CA, Juul SE, Northington F, Bennet L, Gunn AJ. A working model for hypothermic neuroprotection. J Physiol 2018; 596:5641-5654. [PMID: 29660115 DOI: 10.1113/jp274928] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 03/28/2018] [Indexed: 01/04/2023] Open
Abstract
Therapeutic hypothermia significantly improves survival without disability in near-term and full-term newborns with moderate to severe hypoxic-ischaemic encephalopathy. However, hypothermic neuroprotection is incomplete. The challenge now is to find ways to further improve outcomes. One major limitation to progress is that the specific mechanisms of hypothermia are only partly understood. Evidence supports the concept that therapeutic cooling suppresses multiple extracellular death signals, including intracellular pathways of apoptotic and necrotic cell death and inappropriate microglial activation. Thus, the optimal depth of induced hypothermia is that which effectively suppresses the cell death pathways after hypoxia-ischaemia, but without inhibiting recovery of the cellular environment. Thus mild hypothermia needs to be continued until the cell environment has recovered until it can actively support cell survival. This review highlights that key survival cues likely include the inter-related restoration of neuronal activity and growth factor release. This working model suggests that interventions that target overlapping mechanisms, such as anticonvulsants, are unlikely to materially augment hypothermic neuroprotection. We suggest that further improvements are most likely to be achieved with late interventions that maximise restoration of the normal cell environment after therapeutic hypothermia, such as recombinant human erythropoietin or stem cell therapy.
Collapse
Affiliation(s)
- Guido Wassink
- Department of Physiology, University of Auckland, Auckland, New Zealand
| | - Joanne O Davidson
- Department of Physiology, University of Auckland, Auckland, New Zealand
| | | | - Sandra E Juul
- Department of Pediatrics, University of Washington, Seattle, WA, USA
| | - Frances Northington
- Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Laura Bennet
- Department of Physiology, University of Auckland, Auckland, New Zealand
| | - Alistair J Gunn
- Department of Physiology, University of Auckland, Auckland, New Zealand
| |
Collapse
|
20
|
Lin JG, Lin SZ, Lin LH, Wu CC, Tsai WT, Harn HJ, Su LH, Ho WY, Hsieh CJ, Ho TJ. Effects of Moxibustion on the Levels of Insulin-Like Growth Factor 1: A Pilot Study. Cell Transplant 2018; 27:551-556. [PMID: 29692183 PMCID: PMC6038048 DOI: 10.1177/0963689717724795] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Moxibustion (艾灸) is a traditional Chinese medicine therapy performed using Artemisia argyii. Zusanli (足三里, ST36) is an acupoint in the stomach meridian, long associated in ancient Chinese medical practices with the extension of life span when moxibustion is applied to it. The aim of this study was to investigate changes in insulin-like growth factor 1 (IGF-1) levels after application of moxibustion to ST36. Four healthy men and women participated in this 28-day trial and were randomly divided into 2 groups. Group A received moxibustion treatment from days 1 to 14, while group B received moxibustion treatment from days 15 to 28. Blood samples were taken 5 times during this study to measure serum IGF-1 (s-IGF-1) levels. The s-IGF-1 levels increased in both groups after 7 and 14 d of moxibustion therapy (group A: 11.02% [7 d] and 29.65% [14 d]; group B: 169.12% [7 d] and 274.85% [14 d]). After moxibustion therapy had been completed (day 14), s-IGF-1 levels continued to increase in group A (increases on day 21 and day 28 were 53.19% and 61.45%, respectively). There were no adverse events in either group. The s-IGF-1 levels were significantly raised in both groups after 7 and 14 d of moxibustion therapy. Moreover, once therapy had been completed, s-IGF-1 levels continued to increase in group A up to 14 d after the treatment.
Collapse
Affiliation(s)
- Jaung-Geng Lin
- 1 School of Chinese Medicine, China Medical University, Shenyang Shi, Taichung, Taiwan
| | - Shinn-Zong Lin
- 2 Bioinnovation Center, Tzu Chi Foundation, Department of Neurosurgery, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University, Hualien, Taiwan
| | - Lih-Hwa Lin
- 3 Division of Chinese Medicine, An Nan Hospital, China Medical University, Liaoning Sheng, Tainan, Taiwan.,4 Graduate Institute of Chinese Medicine, School of Chinese Medicine, China Medical University, Liaoning Sheng, Taichung, Taiwan
| | - Chun-Chang Wu
- 5 Division of Chinese Medicine, China Medical University Beigang Hospital, Yunlin County, Taiwan
| | - Wan-Ting Tsai
- 5 Division of Chinese Medicine, China Medical University Beigang Hospital, Yunlin County, Taiwan
| | - Horng-Jyh Harn
- 6 Bioinnovation Center, Tzu Chi Foundation, Department of Pathology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University, Hualien, Taiwan
| | - Li-Hui Su
- 7 Department of Nursing, China Medical University Beigang Hospital, Yunlin County, Taiwan
| | - Wen-Yu Ho
- 8 Department of Laboratory Medicine, China Medical University Beigang Hospital, Yunlin County, Taiwan.,9 Department of Medical Technology, Jen-Teh Junior College of Medicine, Nursing and Management, Miaoli County, Taiwan
| | - Che-Jui Hsieh
- 1 School of Chinese Medicine, China Medical University, Shenyang Shi, Taichung, Taiwan
| | - Tsung-Jung Ho
- 1 School of Chinese Medicine, China Medical University, Shenyang Shi, Taichung, Taiwan.,3 Division of Chinese Medicine, An Nan Hospital, China Medical University, Liaoning Sheng, Tainan, Taiwan.,5 Division of Chinese Medicine, China Medical University Beigang Hospital, Yunlin County, Taiwan
| |
Collapse
|
21
|
Tolcos M, Petratos S, Hirst JJ, Wong F, Spencer SJ, Azhan A, Emery B, Walker DW. Blocked, delayed, or obstructed: What causes poor white matter development in intrauterine growth restricted infants? Prog Neurobiol 2017; 154:62-77. [PMID: 28392287 DOI: 10.1016/j.pneurobio.2017.03.009] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 03/17/2017] [Accepted: 03/29/2017] [Indexed: 12/22/2022]
Abstract
Poor white matter development in intrauterine growth restricted (IUGR) babies remains a major, untreated problem in neonatology. New therapies, guided by an understanding of the mechanisms that underlie normal and abnormal oligodendrocyte development and myelin formation, are required. Much of our knowledge of the mechanisms that underlie impaired myelination come from studies in adult demyelinating disease, preterm brain injury, or experimental models of hypoxia-ischemia. However, relatively less is known for IUGR which is surprising because IUGR is a leading cause of perinatal mortality and morbidity, second only to premature birth. IUGR is also a significant risk factor for the later development of cerebral palsy, and is a greater risk compared to some of the more traditionally researched antecedents - asphyxia and inflammation. Recent evidence suggests that the white matter injury and reduced myelination in the brains of some preterm babies is due to impaired maturation of oligodendrocytes thereby resulting in the reduced capacity to synthesize myelin. Therefore, it is not surprising that the hypomyelination observable in the central nervous system of IUGR infants has similarly lead to investigations identifying a delay or blockade in the progress of maturation of oligodendrocytes in these infants. This review will discuss current ideas thought to account for the poor myelination often present in the neonate's brain following IUGR, and discuss novel interventions that are promising as treatments that promote oligodendrocyte maturation, and thereby repair the myelination deficits that otherwise persist into infancy and childhood and lead to neurodevelopmental abnormalities.
Collapse
Affiliation(s)
- Mary Tolcos
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, 3083, Australia.
| | - Steven Petratos
- Department of Medicine, Central Clinical School, Monash University, Prahran, Victoria, 3004, Australia
| | - Jonathan J Hirst
- School of Biomedical Sciences, University of Newcastle, Callaghan, NSW, 2308, Australia
| | - Flora Wong
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, 3168, Australia; Monash Newborn and Monash University, Clayton, Victoria, 3168, Australia
| | - Sarah J Spencer
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, 3083, Australia
| | - Aminath Azhan
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, 3168, Australia
| | - Ben Emery
- Oregon Health and Science University, Portland, OR, 97239-3098, USA
| | - David W Walker
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, 3083, Australia
| |
Collapse
|
22
|
Earnest DJ, Neuendorff N, Coffman J, Selvamani A, Sohrabji F. Sex Differences in the Impact of Shift Work Schedules on Pathological Outcomes in an Animal Model of Ischemic Stroke. Endocrinology 2016; 157:2836-43. [PMID: 27254002 PMCID: PMC4929545 DOI: 10.1210/en.2016-1130] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Circadian clock desynchronization has been implicated in the pathophysiology of cardiovascular disease and related risk factors (eg, obesity, diabetes). Thus, we examined the extent to which circadian desynchronization exacerbates ischemic stroke outcomes and whether its detrimental effects on stroke severity and functional impairments are further modified by biological sex. Circadian entrainment of activity rhythms in all male and female rats was observed during exposure to a fixed light-dark (LD) 12:12 cycle but was severely disrupted when this LD cycle was routinely shifted (12 h advance/5 d) for approximately 7 weeks. In contrast to the regular estrous cycles in fixed LD animals, cyclicity was abolished and persistent estrus was evident in all shifted LD females. The disruption of estrous cyclicity in shifted LD females was associated with a significant increase in serum estradiol levels relative to that observed in fixed LD controls. Circadian rhythm disruption exacerbated stroke outcomes in both shifted LD male and female rats and further amplified sex differences in stroke impairments. In males, but not females, circadian disruption after exposure to the shifted LD cycle was marked by high rates of mortality. In surviving females, circadian desynchronization after exposure to shifted LD cycles produced significant increases in stroke-induced infarct volume and sensorimotor deficits with corresponding decreases in serum IGF-1 levels. These results suggest that circadian rhythm disruption associated with shift work schedules or the irregular nature of our everyday work and/or social environments may interact with other nonmodifiable risk factors such as biological sex to modulate the pathological effects of stroke.
Collapse
Affiliation(s)
- David J Earnest
- Department of Neuroscience and Experimental Therapeutics (D.J.E., N.N., J.C., A.S., F.S.) and Women's Health in Neuroscience Program (A.S., F.S.), Texas A&M Health Science Center, College of Medicine, Bryan, Texas 77807-3260; and Department of Biology (D.J.E.) and Center for Biological Clocks Research (D.J.E.), Texas A&M University, College Station, Texas 77843-3258
| | - Nichole Neuendorff
- Department of Neuroscience and Experimental Therapeutics (D.J.E., N.N., J.C., A.S., F.S.) and Women's Health in Neuroscience Program (A.S., F.S.), Texas A&M Health Science Center, College of Medicine, Bryan, Texas 77807-3260; and Department of Biology (D.J.E.) and Center for Biological Clocks Research (D.J.E.), Texas A&M University, College Station, Texas 77843-3258
| | - Jason Coffman
- Department of Neuroscience and Experimental Therapeutics (D.J.E., N.N., J.C., A.S., F.S.) and Women's Health in Neuroscience Program (A.S., F.S.), Texas A&M Health Science Center, College of Medicine, Bryan, Texas 77807-3260; and Department of Biology (D.J.E.) and Center for Biological Clocks Research (D.J.E.), Texas A&M University, College Station, Texas 77843-3258
| | - Amutha Selvamani
- Department of Neuroscience and Experimental Therapeutics (D.J.E., N.N., J.C., A.S., F.S.) and Women's Health in Neuroscience Program (A.S., F.S.), Texas A&M Health Science Center, College of Medicine, Bryan, Texas 77807-3260; and Department of Biology (D.J.E.) and Center for Biological Clocks Research (D.J.E.), Texas A&M University, College Station, Texas 77843-3258
| | - Farida Sohrabji
- Department of Neuroscience and Experimental Therapeutics (D.J.E., N.N., J.C., A.S., F.S.) and Women's Health in Neuroscience Program (A.S., F.S.), Texas A&M Health Science Center, College of Medicine, Bryan, Texas 77807-3260; and Department of Biology (D.J.E.) and Center for Biological Clocks Research (D.J.E.), Texas A&M University, College Station, Texas 77843-3258
| |
Collapse
|
23
|
Zhang J, Klufas D, Manalo K, Adjepong K, Davidson JO, Wassink G, Bennet L, Gunn AJ, Stopa EG, Liu K, Nishibori M, Stonestreet BS. HMGB1 Translocation After Ischemia in the Ovine Fetal Brain. J Neuropathol Exp Neurol 2016; 75:527-38. [PMID: 27151753 DOI: 10.1093/jnen/nlw030] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Inflammation contributes to the evolution of hypoxic-ischemic (HI) brain injury. High-mobility group box-1 (HMGB1) is a nuclear protein that is translocated from the nucleus and released after ischemia in adult rodents and thereby initiates inflammatory responses. However, there is very little information regarding the effects of HI on HMGB1 in immature brains. To investigate the effects of HI on HMGB1 in the term-equivalent fetal brain, ovine fetuses at 127 days gestation were studied after 30 minutes of carotid occlusion. Groups were sham-control and ischemia with 48 hours and ischemia with 72 hours of reperfusion. By immunohistochemistry, HMGB1 was found to be localized primarily in cell nuclei and partially in cytoplasmic compartments in the cerebral cortex of controls. Ischemia increased the area fraction of neuronal cells with cytoplasmic HMGB1 staining, and Western immunoblot revealed that cytosolic HMGB1 expression increased after ischemia (p < 0.05) and decreased in nuclei in ischemic versus the sham-control brains (p < 0.05). These data indicate that HMGB1 translocates from the nuclear to cytosolic compartments after ischemic brain injury in fetal sheep. This translocation may enable the action of HMGB1 as a proinflammatory cytokine that contributes to HI injury in the developing brain.
Collapse
Affiliation(s)
- Jiyong Zhang
- From the Department of Pediatrics, The Alpert Medical School of Brown University, Women & Infants Hospital of Rhode Island, Providence, Rhode Island (JZ, DK, KM, KA, BSS); Department of Physiology, University of Auckland, Auckland, New Zealand (JOD, GW, LB, AJG); Department of Pathology and Neurosurgery, The Alpert Medical School of Brown University, Rhode Island Hospital, Providence, Rhode Island (EGS); and Dentistry and Pharmaceutical Sciences, Graduate School of Medicine, Okayama University, Okayama, Japan (KL, MN)
| | - Daniel Klufas
- From the Department of Pediatrics, The Alpert Medical School of Brown University, Women & Infants Hospital of Rhode Island, Providence, Rhode Island (JZ, DK, KM, KA, BSS); Department of Physiology, University of Auckland, Auckland, New Zealand (JOD, GW, LB, AJG); Department of Pathology and Neurosurgery, The Alpert Medical School of Brown University, Rhode Island Hospital, Providence, Rhode Island (EGS); and Dentistry and Pharmaceutical Sciences, Graduate School of Medicine, Okayama University, Okayama, Japan (KL, MN)
| | - Karina Manalo
- From the Department of Pediatrics, The Alpert Medical School of Brown University, Women & Infants Hospital of Rhode Island, Providence, Rhode Island (JZ, DK, KM, KA, BSS); Department of Physiology, University of Auckland, Auckland, New Zealand (JOD, GW, LB, AJG); Department of Pathology and Neurosurgery, The Alpert Medical School of Brown University, Rhode Island Hospital, Providence, Rhode Island (EGS); and Dentistry and Pharmaceutical Sciences, Graduate School of Medicine, Okayama University, Okayama, Japan (KL, MN)
| | - Kwame Adjepong
- From the Department of Pediatrics, The Alpert Medical School of Brown University, Women & Infants Hospital of Rhode Island, Providence, Rhode Island (JZ, DK, KM, KA, BSS); Department of Physiology, University of Auckland, Auckland, New Zealand (JOD, GW, LB, AJG); Department of Pathology and Neurosurgery, The Alpert Medical School of Brown University, Rhode Island Hospital, Providence, Rhode Island (EGS); and Dentistry and Pharmaceutical Sciences, Graduate School of Medicine, Okayama University, Okayama, Japan (KL, MN)
| | - Joanne O Davidson
- From the Department of Pediatrics, The Alpert Medical School of Brown University, Women & Infants Hospital of Rhode Island, Providence, Rhode Island (JZ, DK, KM, KA, BSS); Department of Physiology, University of Auckland, Auckland, New Zealand (JOD, GW, LB, AJG); Department of Pathology and Neurosurgery, The Alpert Medical School of Brown University, Rhode Island Hospital, Providence, Rhode Island (EGS); and Dentistry and Pharmaceutical Sciences, Graduate School of Medicine, Okayama University, Okayama, Japan (KL, MN)
| | - Guido Wassink
- From the Department of Pediatrics, The Alpert Medical School of Brown University, Women & Infants Hospital of Rhode Island, Providence, Rhode Island (JZ, DK, KM, KA, BSS); Department of Physiology, University of Auckland, Auckland, New Zealand (JOD, GW, LB, AJG); Department of Pathology and Neurosurgery, The Alpert Medical School of Brown University, Rhode Island Hospital, Providence, Rhode Island (EGS); and Dentistry and Pharmaceutical Sciences, Graduate School of Medicine, Okayama University, Okayama, Japan (KL, MN)
| | - Laura Bennet
- From the Department of Pediatrics, The Alpert Medical School of Brown University, Women & Infants Hospital of Rhode Island, Providence, Rhode Island (JZ, DK, KM, KA, BSS); Department of Physiology, University of Auckland, Auckland, New Zealand (JOD, GW, LB, AJG); Department of Pathology and Neurosurgery, The Alpert Medical School of Brown University, Rhode Island Hospital, Providence, Rhode Island (EGS); and Dentistry and Pharmaceutical Sciences, Graduate School of Medicine, Okayama University, Okayama, Japan (KL, MN)
| | - Alistair J Gunn
- From the Department of Pediatrics, The Alpert Medical School of Brown University, Women & Infants Hospital of Rhode Island, Providence, Rhode Island (JZ, DK, KM, KA, BSS); Department of Physiology, University of Auckland, Auckland, New Zealand (JOD, GW, LB, AJG); Department of Pathology and Neurosurgery, The Alpert Medical School of Brown University, Rhode Island Hospital, Providence, Rhode Island (EGS); and Dentistry and Pharmaceutical Sciences, Graduate School of Medicine, Okayama University, Okayama, Japan (KL, MN)
| | - Edward G Stopa
- From the Department of Pediatrics, The Alpert Medical School of Brown University, Women & Infants Hospital of Rhode Island, Providence, Rhode Island (JZ, DK, KM, KA, BSS); Department of Physiology, University of Auckland, Auckland, New Zealand (JOD, GW, LB, AJG); Department of Pathology and Neurosurgery, The Alpert Medical School of Brown University, Rhode Island Hospital, Providence, Rhode Island (EGS); and Dentistry and Pharmaceutical Sciences, Graduate School of Medicine, Okayama University, Okayama, Japan (KL, MN)
| | - Keyue Liu
- From the Department of Pediatrics, The Alpert Medical School of Brown University, Women & Infants Hospital of Rhode Island, Providence, Rhode Island (JZ, DK, KM, KA, BSS); Department of Physiology, University of Auckland, Auckland, New Zealand (JOD, GW, LB, AJG); Department of Pathology and Neurosurgery, The Alpert Medical School of Brown University, Rhode Island Hospital, Providence, Rhode Island (EGS); and Dentistry and Pharmaceutical Sciences, Graduate School of Medicine, Okayama University, Okayama, Japan (KL, MN)
| | - Masahiro Nishibori
- From the Department of Pediatrics, The Alpert Medical School of Brown University, Women & Infants Hospital of Rhode Island, Providence, Rhode Island (JZ, DK, KM, KA, BSS); Department of Physiology, University of Auckland, Auckland, New Zealand (JOD, GW, LB, AJG); Department of Pathology and Neurosurgery, The Alpert Medical School of Brown University, Rhode Island Hospital, Providence, Rhode Island (EGS); and Dentistry and Pharmaceutical Sciences, Graduate School of Medicine, Okayama University, Okayama, Japan (KL, MN)
| | - Barbara S Stonestreet
- From the Department of Pediatrics, The Alpert Medical School of Brown University, Women & Infants Hospital of Rhode Island, Providence, Rhode Island (JZ, DK, KM, KA, BSS); Department of Physiology, University of Auckland, Auckland, New Zealand (JOD, GW, LB, AJG); Department of Pathology and Neurosurgery, The Alpert Medical School of Brown University, Rhode Island Hospital, Providence, Rhode Island (EGS); and Dentistry and Pharmaceutical Sciences, Graduate School of Medicine, Okayama University, Okayama, Japan (KL, MN).
| |
Collapse
|
24
|
Davidson JO, Yuill CA, Zhang FG, Wassink G, Bennet L, Gunn AJ. Extending the duration of hypothermia does not further improve white matter protection after ischemia in term-equivalent fetal sheep. Sci Rep 2016; 6:25178. [PMID: 27121655 PMCID: PMC4848549 DOI: 10.1038/srep25178] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 04/12/2016] [Indexed: 11/29/2022] Open
Abstract
A major challenge in modern neonatal care is to further improve outcomes after therapeutic hypothermia for hypoxic ischemic encephalopathy. In this study we tested whether extending the duration of cooling might reduce white matter damage. Term-equivalent fetal sheep (0.85 gestation) received either sham ischemia followed by normothermia (n = 8) or 30 minutes of bilateral carotid artery occlusion followed by three days of normothermia (n = 8), three days of hypothermia (n = 8) or five days of hypothermia (n = 8) started three hours after ischemia. Histology was assessed 7 days after ischemia. Ischemia was associated with loss of myelin basic protein (MBP) and Olig-2 positive oligodendrocytes and increased Iba-1-positive microglia compared to sham controls (p < 0.05). Three days and five days of hypothermia were associated with a similar, partial improvement in MBP and numbers of oligodendrocytes compared to ischemia-normothermia (p < 0.05). Both hypothermia groups had reduced microglial activation compared to ischemia-normothermia (p < 0.05). In the ischemia-five-day hypothermia group, but not ischemia-three-day, numbers of microglia remained higher than in sham controls (p < 0.05). In conclusion, delayed cerebral hypothermia partially protected white matter after global cerebral ischemia in fetal sheep. Extending cooling from 3 to 5 days did not further improve outcomes, and may be associated with greater numbers of residual microglia.
Collapse
Affiliation(s)
- Joanne O Davidson
- Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Caroline A Yuill
- Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Frank G Zhang
- Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Guido Wassink
- Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Laura Bennet
- Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Alistair J Gunn
- Department of Physiology, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
25
|
Davidson JO, Wassink G, van den Heuij LG, Bennet L, Gunn AJ. Therapeutic Hypothermia for Neonatal Hypoxic-Ischemic Encephalopathy - Where to from Here? Front Neurol 2015; 6:198. [PMID: 26441818 PMCID: PMC4568393 DOI: 10.3389/fneur.2015.00198] [Citation(s) in RCA: 142] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 08/31/2015] [Indexed: 11/13/2022] Open
Abstract
Hypoxia-ischemia before or around the time of birth occurs in approximately 2/1000 live births and is associated with a high risk of death or lifelong disability. Therapeutic hypothermia is now well established as standard treatment for infants with moderate to severe hypoxic-ischemic encephalopathy but is only partially effective. There is compelling preclinical and clinical evidence that hypothermia is most protective when it is started as early as possible after hypoxia-ischemia. Further improvements in outcome from therapeutic hypothermia are very likely to arise from strategies to reduce the delay before starting treatment of affected infants. In this review, we examine evidence that current protocols are reasonably close to the optimal depth and duration of cooling, but that the optimal rate of rewarming after hypothermia is unclear. The potential for combination treatments to augment hypothermic neuroprotection has considerable promise, particularly with endogenous targets such as melatonin and erythropoietin, and noble gases such as xenon. We dissect the critical importance of preclinical studies using realistic delays in treatment and clinically relevant cooling protocols when examining combination treatment, and that for many strategies overlapping mechanisms of action can substantially attenuate any effects.
Collapse
Affiliation(s)
- Joanne O Davidson
- The Department of Physiology, The University of Auckland , Auckland , New Zealand
| | - Guido Wassink
- The Department of Physiology, The University of Auckland , Auckland , New Zealand
| | | | - Laura Bennet
- The Department of Physiology, The University of Auckland , Auckland , New Zealand
| | - Alistair J Gunn
- The Department of Physiology, The University of Auckland , Auckland , New Zealand
| |
Collapse
|
26
|
Back SA. Brain Injury in the Preterm Infant: New Horizons for Pathogenesis and Prevention. Pediatr Neurol 2015; 53:185-92. [PMID: 26302698 PMCID: PMC4550810 DOI: 10.1016/j.pediatrneurol.2015.04.006] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Revised: 03/24/2015] [Accepted: 04/12/2015] [Indexed: 10/23/2022]
Abstract
Preterm neonates are surviving with a milder spectrum of motor and cognitive disabilities that appear to be related to widespread disturbances in cell maturation that target cerebral gray and white matter. Whereas the preterm brain was previously at high risk for destructive lesions, preterm survivors now commonly display less severe injury that is associated with aberrant regeneration and repair responses that result in reduced cerebral growth. Impaired cerebral white matter growth is related to myelination disturbances that are initiated by acute death of premyelinating oligodendrocytes, but are followed by rapid regeneration of premyelinating oligodendrocytes that fail to normally mature to myelinating cells. Although immature neurons are more resistant to cell death than mature neurons, they display widespread disturbances in maturation of their dendritic arbors and synapses, which further contributes to impaired cerebral growth. Thus, even more mild cerebral injury involves disrupted repair mechanisms in which neurons and premyelinating oligodendrocytes fail to fully mature during a critical window in development of neural circuitry. These recently recognized distinct forms of cerebral gray and white matter dysmaturation raise new diagnostic challenges and suggest new therapeutic strategies to promote brain growth and repair.
Collapse
Affiliation(s)
- Stephen A. Back
- Departments of Pediatrics and Neurology, Oregon Health & Science University, Portland, Oregon, U.S.A
| |
Collapse
|
27
|
Rong Z, Pan R, Chang L, Lee W. Combination treatment with ethyl pyruvate and IGF-I exerts neuroprotective effects against brain injury in a rat model of neonatal hypoxic-ischemic encephalopathy. Int J Mol Med 2015; 36:195-203. [PMID: 25999282 PMCID: PMC4494588 DOI: 10.3892/ijmm.2015.2219] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 05/11/2015] [Indexed: 01/28/2023] Open
Abstract
Neonatal hypoxic-ischemic (HI) brain injury causes severe brain damage in newborns. Following HI injury, rapidly accumulating oxidants injure neurons and interrupt ongoing developmental processes. The antioxidant, sodium pyruvate, has been shown to reduce neuronal injury in neonatal rats under conditions of oxygen glucose deprivation (OGD) and HI injury. In this study, we evaluated the effects of ethyl pyruvate (EP) and insulin-like growth factor-I (IGF-I) alone or in combination in a similar setting. For this purpose, we used an in vitro model involving primary neonatal rat cortical neurons subjected to OGD for 2.5 h and an in vivo model involving unilateral carotid ligation in rats on post-natal day 7 with exposure to 8% hypoxia for 2.5 h. The cultured neurons were examined by lactate dehydrogenase (LDH) and cell viability assays. For the in vivo experiments, behavioral development was evaluated by the foot fault test at 4 weeks of recovery. 2,3,5-Triphenyltetrazolium chloride monohydrate and cresyl violet staining were used to evaluate HI injury. The injured neurons were Fluoro-Jade B-labeled, new neuroprecursors were double labeled with bromodeoxyuridine (BrdU) and doublecortin, new mature neurons were BrdU-labeled and neuronal nuclei were labeled by immunofluorescence. Under conditions of OGD, the LDH levels increased and neuronal viability decreased. Treatment with 0.5 mM EP or 25 ng/ml IGF-I protected the neurons (P<0.05), exerting additive effects. Similarly, either the early administration of EP or delayed treatment with IGF-I protected the neonatal rat brains against HI injury and improved neurological performance and these effects were also additive. This effect may be the result of reduced neuronal injury, and enhanced neurogenesis and maturation. On the whole, our findings demonstrate that the combination of the early administration of EP with delayed treatment with IGF-I exerts neuroprotective effects against HI injury in neonatal rat brains.
Collapse
Affiliation(s)
- Zhihui Rong
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Rui Pan
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Liwen Chang
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Weihua Lee
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
28
|
Larpthaveesarp A, Ferriero DM, Gonzalez FF. Growth factors for the treatment of ischemic brain injury (growth factor treatment). Brain Sci 2015; 5:165-77. [PMID: 25942688 PMCID: PMC4493462 DOI: 10.3390/brainsci5020165] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 04/13/2015] [Accepted: 04/21/2015] [Indexed: 12/11/2022] Open
Abstract
In recent years, growth factor therapy has emerged as a potential treatment for ischemic brain injury. The efficacy of therapies that either directly introduce or stimulate local production of growth factors and their receptors in damaged brain tissue has been tested in a multitude of models for different Central Nervous System (CNS) diseases. These growth factors include erythropoietin (EPO), vascular endothelial growth factor (VEGF), brain-derived neurotrophic factor (BDNF), and insulin-like growth factor (IGF-1), among others. Despite the promise shown in animal models, the particular growth factors that should be used to maximize both brain protection and repair, and the therapeutic critical period, are not well defined. We will review current pre-clinical and clinical evidence for growth factor therapies in treating different causes of brain injury, as well as issues to be addressed prior to application in humans.
Collapse
Affiliation(s)
- Amara Larpthaveesarp
- Department of Pediatrics, University of California, San Francisco, CA 94158, USA.
| | - Donna M Ferriero
- Departments of Pediatrics and Neurology, University of California, San Francisco, CA 94158, USA.
| | - Fernando F Gonzalez
- Department of Pediatrics, University of California, San Francisco, CA 94158, USA.
| |
Collapse
|
29
|
Guan J, Harris P, Brimble M, Lei Y, Lu J, Yang Y, Gunn AJ. The role for IGF-1-derived small neuropeptides as a therapeutic target for neurological disorders. Expert Opin Ther Targets 2015; 19:785-93. [PMID: 25652713 DOI: 10.1517/14728222.2015.1010514] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
INTRODUCTION Exogenous IGF-1 protects the brain from ischemic injury and improves function. However, its clinical application to neurological disorders is limited by its large molecular size, poor central uptake and mitogenic potential. AREAS COVERED In this review, the authors have discussed the efficacy, pharmacokinetics and mechanisms of IGF-1 derivatives on protecting acute brain injury, preventing memory impairment and improving recovery from neurological degenerative conditions evaluated in various animal models. We have included natural metabolites of IGF-1, glycine-proline-glutamate (GPE), cleaved from N-terminal IGF-1 and cyclic glycine-proline (cGP) as well as the structural analogues of GPE and cGP, glycine-2-methyl-proline-glutamate and cyclo-l-glycyl-l-2-allylproline, respectively. In addition, the regulatory role for cGP in bioavailability of IGF-1 has also been discussed. EXPERT OPINION These small neuropeptides provide effective neuroprotection by offering an improved pharmacokinetic profile and more practical route of administration compared with IGF-1 administration. Developing modified neuropeptides to overcome the limitations of their endogenous counterparts represents a novel strategy of pharmaceutical discovery for neurological disorders. The mechanism of action may involve a regulation of IGF-1 bioavailability.
Collapse
Affiliation(s)
- Jian Guan
- University of Auckland, Liggins Institute , Private Bag 92019, Auckland , New Zealand +64 93 737 599 ext. 86134 ; +64 93 082 385 ;
| | | | | | | | | | | | | |
Collapse
|
30
|
Sohrabji F. Estrogen-IGF-1 interactions in neuroprotection: ischemic stroke as a case study. Front Neuroendocrinol 2015; 36:1-14. [PMID: 24882635 PMCID: PMC4247812 DOI: 10.1016/j.yfrne.2014.05.003] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2013] [Revised: 05/15/2014] [Accepted: 05/16/2014] [Indexed: 12/25/2022]
Abstract
The steroid hormone 17b-estradiol and the peptide hormone insulin-like growth factor (IGF)-1 independently exert neuroprotective actions in neurologic diseases such as stroke. Only a few studies have directly addressed the interaction between the two hormone systems, however, there is a large literature that indicates potentially greater interactions between the 17b-estradiol and IGF-1 systems. The present review focuses on key issues related to this interaction including IGF-1 and sex differences and common activation of second messenger systems. Using ischemic stroke as a case study, this review also focuses on independent and cooperative actions of estrogen and IGF-1 on neuroprotection, blood brain barrier integrity, angiogenesis, inflammation and post-stroke epilepsy. Finally, the review also focuses on the astrocyte, a key mediator of post stroke repair, as a local source of 17b-estradiol and IGF-1. This review thus highlights areas where significant new research is needed to clarify the interactions between these two neuroprotectants.
Collapse
Affiliation(s)
- Farida Sohrabji
- Women's Health in Neuroscience Program, Neuroscience and Experimental Therapeutics, TAMHSC College of Medicine, Bryan, TX 77807, United States.
| |
Collapse
|
31
|
Elitt CM, Rosenberg PA. The challenge of understanding cerebral white matter injury in the premature infant. Neuroscience 2014; 276:216-38. [PMID: 24838063 DOI: 10.1016/j.neuroscience.2014.04.038] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Revised: 04/15/2014] [Accepted: 04/15/2014] [Indexed: 12/18/2022]
Abstract
White matter injury in the premature infant leads to motor and more commonly behavioral and cognitive problems that are a tremendous burden to society. While there has been much progress in understanding unique vulnerabilities of developing oligodendrocytes over the past 30years, there remain no proven therapies for the premature infant beyond supportive care. The lack of translational progress may be partially explained by the challenge of developing relevant animal models when the etiology remains unclear, as is the case in this disorder. There has been an emphasis on hypoxia-ischemia and infection/inflammation as upstream etiologies, but less consideration of other contributory factors. This review highlights the evolution of white matter pathology in the premature infant, discusses the prevailing proposed etiologies, critically analyzes a sampling of common animal models and provides detailed support for our hypothesis that nutritional and hormonal deprivation may be additional factors playing critical and overlooked roles in white matter pathology in the premature infant.
Collapse
Affiliation(s)
- C M Elitt
- Department of Neurology and the F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA; Program in Neuroscience, Harvard Medical School, Boston, MA 02115, USA
| | - P A Rosenberg
- Department of Neurology and the F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA; Program in Neuroscience, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
32
|
Back SA, Miller SP. Brain injury in premature neonates: A primary cerebral dysmaturation disorder? Ann Neurol 2014; 75:469-86. [PMID: 24615937 PMCID: PMC5989572 DOI: 10.1002/ana.24132] [Citation(s) in RCA: 254] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Revised: 03/04/2014] [Accepted: 03/05/2014] [Indexed: 12/11/2022]
Abstract
With advances in neonatal care, preterm neonates are surviving with an evolving constellation of motor and cognitive disabilities that appear to be related to widespread cellular maturational disturbances that target cerebral gray and white matter. Whereas preterm infants were previously at high risk for destructive brain lesions that resulted in cystic white matter injury and secondary cortical and subcortical gray matter degeneration, contemporary cohorts of preterm survivors commonly display less severe injury that does not appear to involve pronounced glial or neuronal loss. Nevertheless, these milder forms of injury are also associated with reduced cerebral growth. Recent human and experimental studies support that impaired cerebral growth is related to disparate responses in gray and white matter. Myelination disturbances in cerebral white matter are related to aberrant regeneration and repair responses to acute death of premyelinating late oligodendrocyte progenitors (preOLs). In response to preOL death, early oligodendrocyte progenitors rapidly proliferate and differentiate, but the regenerated preOLs fail to normally mature to myelinating cells required for white matter growth. Although immature neurons appear to be more resistant to cell death from hypoxia-ischemia than glia, they display widespread disturbances in maturation of their dendritic arbors, which further contribute to impaired cerebral growth. These complex and disparate responses of neurons and preOLs thus result in large numbers of cells that fail to fully mature during a critical window in development of neural circuitry. These recently recognized forms of cerebral gray and white matter dysmaturation raise new diagnostic challenges and suggest new therapeutic directions centered on reversal of the processes that promote dysmaturation.
Collapse
Affiliation(s)
- Stephen A Back
- Departments of Pediatrics, Oregon Health and Science University, Portland; Departments of Neurology, Oregon Health and Science University, Portland
| | | |
Collapse
|
33
|
Back SA, Rosenberg PA. Pathophysiology of glia in perinatal white matter injury. Glia 2014; 62:1790-815. [PMID: 24687630 DOI: 10.1002/glia.22658] [Citation(s) in RCA: 153] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 02/13/2014] [Accepted: 02/27/2014] [Indexed: 12/12/2022]
Abstract
Injury to the preterm brain has a particular predilection for cerebral white matter. White matter injury (WMI) is the most common cause of brain injury in preterm infants and a major cause of chronic neurological morbidity including cerebral palsy. Factors that predispose to WMI include cerebral oxygenation disturbances and maternal-fetal infection. During the acute phase of WMI, pronounced oxidative damage occurs that targets late oligodendrocyte progenitors (pre-OLs). The developmental predilection for WMI to occur during prematurity appears to be related to both the timing of appearance and regional distribution of susceptible pre-OLs that are vulnerable to a variety of chemical mediators including reactive oxygen species, glutamate, cytokines, and adenosine. During the chronic phase of WMI, the white matter displays abberant regeneration and repair responses. Early OL progenitors respond to WMI with a rapid robust proliferative response that results in a several fold regeneration of pre-OLs that fail to terminally differentiate along their normal developmental time course. Pre-OL maturation arrest appears to be related in part to inhibitory factors that derive from reactive astrocytes in chronic lesions. Recent high field magnetic resonance imaging (MRI) data support that three distinct forms of chronic WMI exist, each of which displays unique MRI and histopathological features. These findings suggest the possibility that therapies directed at myelin regeneration and repair could be initiated early after WMI and monitored over time. These new mechanisms of acute and chronic WMI provide access to a variety of new strategies to prevent or promote repair of WMI in premature infants.
Collapse
Affiliation(s)
- Stephen A Back
- Department of Pediatrics, Oregon Health and Science University, Portland, Oregon; Department of Neurology, Oregon Health and Science University, Portland, Oregon
| | | |
Collapse
|
34
|
Wassink G, Gunn ER, Drury PP, Bennet L, Gunn AJ. The mechanisms and treatment of asphyxial encephalopathy. Front Neurosci 2014; 8:40. [PMID: 24578682 PMCID: PMC3936504 DOI: 10.3389/fnins.2014.00040] [Citation(s) in RCA: 140] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2013] [Accepted: 02/12/2014] [Indexed: 11/13/2022] Open
Abstract
Acute post-asphyxial encephalopathy occurring around the time of birth remains a major cause of death and disability. The recent seminal insight that allows active neuroprotective treatment is that even after profound asphyxia (the “primary” phase), many brain cells show initial recovery from the insult during a short “latent” phase, typically lasting approximately 6 h, only to die hours to days later after a “secondary” deterioration characterized by seizures, cytotoxic edema, and progressive failure of cerebral oxidative metabolism. Although many of these secondary processes are potentially injurious, they appear to be primarily epiphenomena of the “execution” phase of cell death. Animal and human studies designed around this conceptual framework have shown that moderate cerebral hypothermia initiated as early as possible but before the onset of secondary deterioration, and continued for a sufficient duration to allow the secondary deterioration to resolve, has been associated with potent, long-lasting neuroprotection. Recent clinical trials show that while therapeutic hypothermia significantly reduces morbidity and mortality, many babies still die or survive with disabilities. The challenge for the future is to find ways of improving the effectiveness of treatment. In this review, we will dissect the known mechanisms of hypoxic-ischemic brain injury in relation to the known effects of hypothermic neuroprotection.
Collapse
Affiliation(s)
- Guido Wassink
- Fetal Physiology and Neuroscience Team, Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland Auckland, New Zealand
| | - Eleanor R Gunn
- Fetal Physiology and Neuroscience Team, Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland Auckland, New Zealand
| | - Paul P Drury
- Fetal Physiology and Neuroscience Team, Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland Auckland, New Zealand
| | - Laura Bennet
- Fetal Physiology and Neuroscience Team, Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland Auckland, New Zealand
| | - Alistair J Gunn
- Fetal Physiology and Neuroscience Team, Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland Auckland, New Zealand
| |
Collapse
|
35
|
Sohrabji F, Williams M. Stroke neuroprotection: oestrogen and insulin-like growth factor-1 interactions and the role of microglia. J Neuroendocrinol 2013; 25:1173-81. [PMID: 23763366 PMCID: PMC5630268 DOI: 10.1111/jne.12059] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Revised: 05/30/2013] [Accepted: 06/09/2013] [Indexed: 12/25/2022]
Abstract
Oestrogen has been shown to be neuroprotective for stroke and other neural injury models. Oestrogen promotes a neuroprotective phenotype through myriad actions, including stimulating neurogenesis, promoting neuronal differentiation and survival, suppressing neuroinflammation and maintaining the integrity of the blood-brain barrier. At the molecular level, oestrogen directly modulates genes that are beneficial for repair and regeneration via the canonical oestrogen receptor. Increasingly, evidence indicates that oestrogen acts in concert with growth factors to initiate neuroprotection. Oestrogen and insulin-like growth factor (IGF)-1 act cooperatively to influence cell survival, and combined steroid hormone/growth factor interaction has been well documented in the context of neurones and astrocytes. Here, we summarise the evidence that oestrogen-mediated neuroprotection is critically dependent on IGF-1 signalling, and specifically focus on microglia as the source of IGF-1 and the locus of oestrogen-IGF-1 interactions in stroke neuroprotection.
Collapse
Affiliation(s)
- F Sohrabji
- Women's Health in Neuroscience Program, Neuroscience and Experimental Therapeutics, TAMHSC College of Medicine, Bryan, TX, USA
| | | |
Collapse
|
36
|
Dani C, Poggi C, Bresci C, Corsini I, Frosini S, Pratesi S. Early fresh-frozen plasma transfusion decreases the risk of retinopathy of prematurity. Transfusion 2013; 54:1002-7. [PMID: 24117975 DOI: 10.1111/trf.12432] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Revised: 07/30/2013] [Accepted: 07/30/2013] [Indexed: 11/27/2022]
Abstract
BACKGROUND Insulin-like growth factor 1 (IGF-1) and IGF-binding protein-3 (IGFBP-3) play a main role in the pathogenesis of retinopathy of prematurity (ROP). Fresh-frozen plasma (FFP) from adult donors may be an actual source of IGF-1 and IGFBP-3 because it contains higher concentrations. The objective was to evaluate whether FFP transfusions can decrease the occurrence of ROP in a cohort of preterm infants. STUDY DESIGN AND METHODS We retrospectively analyzed data from 218 infants with gestational age of less than 29 weeks who either received FFP or did not and correlated this procedure to the development of any grade of ROP. RESULTS Logistic regression analysis demonstrated that two or more transfusions of FFP was effective in decreasing the risk of development of any grade of ROP (relative risk, 0.46; 95% confidence interval, 0.23-0.93). Other factors that affected the risk of ROP were gestational age, birthweight, antenatal steroid treatment, FiO2 of at least 0.40, mechanical ventilation, and sepsis. CONCLUSIONS We found that two or more transfusions of FFP in the first week of life decrease the risk of developing any grade of ROP in preterm infants with gestational age of less than 29 weeks.
Collapse
Affiliation(s)
- Carlo Dani
- Department of Neurosciences, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | | | | | | | | | | |
Collapse
|
37
|
Sonntag WE, Deak F, Ashpole N, Toth P, Csiszar A, Freeman W, Ungvari Z. Insulin-like growth factor-1 in CNS and cerebrovascular aging. Front Aging Neurosci 2013; 5:27. [PMID: 23847531 PMCID: PMC3698444 DOI: 10.3389/fnagi.2013.00027] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Accepted: 06/14/2013] [Indexed: 12/26/2022] Open
Abstract
Insulin-like growth factor-1 (IGF-1) is an important anabolic hormone that decreases with age. In the past two decades, extensive research has determined that the reduction in IGF-1 is an important component of the age-related decline in cognitive function in multiple species including humans. Deficiency in circulating IGF-1 results in impairment in processing speed and deficiencies in both spatial and working memory. Replacement of IGF-1 or factors that increase IGF-1 to old animals and humans reverses many of these cognitive deficits. Despite the overwhelming evidence for IGF-1 as an important neurotrophic agent, the specific mechanisms through which IGF-1 acts have remained elusive. Recent evidence indicates that IGF-1 is both produced by and has important actions on the cerebrovasculature as well as neurons and glia. Nevertheless, the specific regulation and actions of brain- and vascular-derived IGF-1 is poorly understood. The diverse effects of IGF-1 discovered thus far reveal a complex endocrine and paracrine system essential for integrating many of the functions necessary for brain health. Identification of the mechanisms of IGF-1 actions will undoubtedly provide critical insight into regulation of brain function in general and the causes of cognitive decline with age.
Collapse
Affiliation(s)
- William E Sonntag
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center Oklahoma City, OK, USA
| | | | | | | | | | | | | |
Collapse
|
38
|
Gutiérrez-Fernández M, Fuentes B, Rodríguez-Frutos B, Ramos-Cejudo J, Vallejo-Cremades MT, Díez-Tejedor E. Trophic factors and cell therapy to stimulate brain repair after ischaemic stroke. J Cell Mol Med 2012; 16:2280-90. [PMID: 22452968 PMCID: PMC3823421 DOI: 10.1111/j.1582-4934.2012.01575.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2011] [Accepted: 03/21/2012] [Indexed: 12/11/2022] Open
Abstract
Brain repair involves a compendium of natural mechanisms that are activated following stroke. From a therapeutic viewpoint, reparative therapies that encourage cerebral plasticity are needed. In the last years, it has been demonstrated that modulatory treatments for brain repair such as trophic factor- and stem cell-based therapies can promote neurogenesis, gliogenesis, oligodendrogenesis, synaptogenesis and angiogenesis, all of which having a beneficial impact on infarct volume, cell death and, finally, and most importantly, on the functional recovery. However, even when promising results have been obtained in a wide range of experimental animal models and conditions these preliminary results have not yet demonstrated their clinical efficacy. Here, we focus on brain repair modulatory treatments for ischaemic stroke, that use trophic factors, drugs with trophic effects and stem cell therapy. Important and still unanswered questions for translational research ranging from experimental animal models to recent and ongoing clinical trials are reviewed here.
Collapse
Affiliation(s)
- María Gutiérrez-Fernández
- Department of Neurology and Stroke Centre, La Paz University Hospital Neuroscience Area of IdiPAZ (Health Research Institute) Autónoma University of MadridMadrid, Spain
| | - Blanca Fuentes
- Neuroscience and Cerebrovascular Research Laboratory, La Paz University Hospital Neuroscience Area of IdiPAZ (Health Research Institute) Autónoma University of MadridMadrid, Spain
| | - Berta Rodríguez-Frutos
- Department of Neurology and Stroke Centre, La Paz University Hospital Neuroscience Area of IdiPAZ (Health Research Institute) Autónoma University of MadridMadrid, Spain
| | - Jaime Ramos-Cejudo
- Department of Neurology and Stroke Centre, La Paz University Hospital Neuroscience Area of IdiPAZ (Health Research Institute) Autónoma University of MadridMadrid, Spain
| | - María Teresa Vallejo-Cremades
- Department of Neurology and Stroke Centre, La Paz University Hospital Neuroscience Area of IdiPAZ (Health Research Institute) Autónoma University of MadridMadrid, Spain
| | - Exuperio Díez-Tejedor
- Department of Neurology and Stroke Centre, La Paz University Hospital Neuroscience Area of IdiPAZ (Health Research Institute) Autónoma University of MadridMadrid, Spain
- Neuroscience and Cerebrovascular Research Laboratory, La Paz University Hospital Neuroscience Area of IdiPAZ (Health Research Institute) Autónoma University of MadridMadrid, Spain
| |
Collapse
|
39
|
Ungvari Z, Csiszar A. The emerging role of IGF-1 deficiency in cardiovascular aging: recent advances. J Gerontol A Biol Sci Med Sci 2012; 67:599-610. [PMID: 22451468 DOI: 10.1093/gerona/gls072] [Citation(s) in RCA: 114] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
This review focuses on cardiovascular protective effects of insulin-like growth factor (IGF)-1, provides a landscape of molecular mechanisms involved in cardiovascular alterations in patients and animal models with congenital and adult-onset IGF-1 deficiency, and explores the link between age-related IGF-1 deficiency and the molecular, cellular, and functional changes that occur in the cardiovascular system during aging. Microvascular protection conferred by endocrine and paracrine IGF-1 signaling, its implications for the pathophysiology of cardiac failure and vascular cognitive impairment, and the role of impaired cellular stress resistance in cardiovascular aging considered here are based on emerging knowledge of the effects of IGF-1 on Nrf2-driven antioxidant response.
Collapse
Affiliation(s)
- Zoltan Ungvari
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1303, Oklahoma City, OK 73104, USA.
| | | |
Collapse
|
40
|
Selvamani A, Sathyan P, Miranda RC, Sohrabji F. An antagomir to microRNA Let7f promotes neuroprotection in an ischemic stroke model. PLoS One 2012; 7:e32662. [PMID: 22393433 PMCID: PMC3290559 DOI: 10.1371/journal.pone.0032662] [Citation(s) in RCA: 178] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2011] [Accepted: 01/28/2012] [Indexed: 11/23/2022] Open
Abstract
We previously showed that middle-aged female rats sustain a larger infarct following experimental stroke as compared to younger female rats, and paradoxically, estrogen treatment to the older group is neurotoxic. Plasma and brain insulin-like growth factor-1 (IGF-1) levels decrease with age. However, IGF-1 infusion following stroke, prevents estrogen neurotoxicity in middle-aged female rats. IGF1 is neuroprotective and well tolerated, but also has potentially undesirable side effects. We hypothesized that microRNAs (miRNAs) that target the IGF-1 signaling family for translation repression could be alternatively suppressed to promote IGF-1-like neuroprotection. Here, we report that two conserved IGF pathway regulatory microRNAs, Let7f and miR1, can be inhibited to mimic and even extend the neuroprotection afforded by IGF-1. Anti-mir1 treatment, as late as 4 hours following ischemia, significantly reduced cortical infarct volume in adult female rats, while anti-Let7 robustly reduced both cortical and striatal infarcts, and preserved sensorimotor function and interhemispheric neural integration. No neuroprotection was observed in animals treated with a brain specific miRNA unrelated to IGF-1 (anti-miR124). Remarkably, anti-Let7f was only effective in intact females but not males or ovariectomized females indicating that the gonadal steroid environment critically modifies miRNA action. Let7f is preferentially expressed in microglia in the ischemic hemisphere and confirmed in ex vivo cultures of microglia obtained from the cortex. While IGF-1 was undetectable in microglia harvested from the non-ischemic hemisphere, IGF-1 was expressed by microglia obtained from the ischemic cortex and was further elevated by anti-Let7f treatment. Collectively these data support a novel miRNA-based therapeutic strategy for neuroprotection following stroke.
Collapse
Affiliation(s)
| | | | | | - Farida Sohrabji
- Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center College of Medicine, College Station, Texas, United States of America
- * E-mail:
| |
Collapse
|
41
|
Kaur C, Rathnasamy G, Ling EA. Roles of activated microglia in hypoxia induced neuroinflammation in the developing brain and the retina. J Neuroimmune Pharmacol 2012; 8:66-78. [PMID: 22367679 DOI: 10.1007/s11481-012-9347-2] [Citation(s) in RCA: 147] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2011] [Accepted: 02/09/2012] [Indexed: 12/14/2022]
Abstract
Amoeboid microglial cells (AMCs) in the developing brain display surface receptors and antigens shared by the monocyte-derived tissue macrophages. Activation of AMCs in the perinatal brain has been associated with periventricular white matter damage in hypoxic-ischemic conditions. The periventricular white matter, where the AMCs preponderate, is selectively vulnerable to hypoxia as manifested by death of premyelinating oligodendrocytes and degeneration of axons leading to neonatal mortality and long-term neurodevelopmental deficits. AMCs respond vigorously to hypoxia by producing excess amounts of inflammatory cytokines e.g. the tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) along with glutamate, nitric oxide (NO) and reactive oxygen species which collectively cause oligodendrocyte death, axonal degeneration as well as disruption of the immature blood brain barrier. A similar phenomenon is observed in the hypoxic developing cerebellum in which activated AMCs induced Purkinje neuronal death through production of TNF-α and IL-1β via their respective receptors. Hypoxia is also implicated in retinopathy of prematurity in which activation of AMCs has been shown to cause retinal ganglion cell death through production of TNF-α and IL-1β and NO. Because AMCs play a pivotal role in hypoxic injuries in the developing brain affecting both neurons and oligodendrocytes, a fuller understanding of the underlying molecular mechanisms of microglial activation under such conditions would be desirable for designing of a novel therapeutic strategy for management of hypoxic damage.
Collapse
Affiliation(s)
- Charanjit Kaur
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, 4 Medical Drive, MD10, Singapore 117597, Singapore.
| | | | | |
Collapse
|
42
|
Cai Z, Fan LW, Lin S, Pang Y, Rhodes PG. Intranasal administration of insulin-like growth factor-1 protects against lipopolysaccharide-induced injury in the developing rat brain. Neuroscience 2011; 194:195-207. [PMID: 21840378 DOI: 10.1016/j.neuroscience.2011.08.003] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2011] [Revised: 08/01/2011] [Accepted: 08/01/2011] [Indexed: 11/27/2022]
Abstract
Our previous studies show that insulin-like growth factor-1 (IGF-1) can either protect against or increase lipopolysaccharide (LPS)-induced damage in the developing brain, depending on the dose, when it is co-administered with LPS through intracerebral injection. To further explore effects of IGF-1 on central inflammation associated brain injury, IGF-1 was administered through intranasal infusion in the current study. Postnatal day 5 (P5) rats were exposed to LPS at a dose of 1 μg/g body weight or sterile saline through intracerebral injection. Recombinant human insulin-like growth factor-1 (rhIGF-1) at a dose of 50 μg/pup or vehicle was administered intranasally 1 or 2 h after the LPS injection. Neonatal LPS exposure resulted in oligodendrocyte (OL) and white matter injury in the P6 or P21 rat brain. The damages include dilatation of lateral ventricles, pyknotic cell death, loss of OL progenitor cells and mature OLs in the cingulum area, and impairment of myelination at the corpus callosum area. Neurological dysfunctions were observed in juvenile rats with neonatal LPS exposure. Intranasal IGF-1 treatment at either 1 or 2 h after LPS exposure significantly attenuated LPS-induced brain injury and improved some behavioral deficits. Intranasal IGF-1 treatment also reduced infiltration of polymorphonuclear (PMN) leukocytes and activation of microglia in the rat brain 24 h after LPS exposure, but it did not prevent the elevation in concentrations of interleukin-1β (IL-1β) and tumor necrosis factor alpha (TNFα) in the LPS-exposed rat brain during the first 24 h. This is an indication that direct anti-inflammation might not be the primary mechanism for the protection of IGF-1, and other mechanisms, such as anti-apoptotic effects, are likely involved in its protective effects.
Collapse
Affiliation(s)
- Z Cai
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA.
| | | | | | | | | |
Collapse
|
43
|
George S, Bennet L, Weaver-Mikaere L, Fraser M, Bouwmans J, Mathai S, Skinner S, Gunn A. White Matter Protection with Insulin-Like Growth Factor 1 and Hypothermia Is Not Additive after Severe Reversible Cerebral Ischemia in Term Fetal Sheep. Dev Neurosci 2011; 33:280-7. [DOI: 10.1159/000329923] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2010] [Accepted: 06/06/2011] [Indexed: 12/13/2022] Open
|
44
|
Mi S, Lee X, Hu Y, Ji B, Shao Z, Yang W, Huang G, Walus L, Rhodes K, Gong BJ, Miller RH, Pepinsky RB. Death receptor 6 negatively regulates oligodendrocyte survival, maturation and myelination. Nat Med 2011; 17:816-21. [PMID: 21725297 DOI: 10.1038/nm.2373] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2010] [Accepted: 04/06/2011] [Indexed: 12/11/2022]
Abstract
Survival and differentiation of oligodendrocytes are important for the myelination of central nervous system (CNS) axons during development and crucial for myelin repair in CNS demyelinating diseases such as multiple sclerosis. Here we show that death receptor 6 (DR6) is a negative regulator of oligodendrocyte maturation. DR6 is expressed strongly in immature oligodendrocytes and weakly in mature myelin basic protein (MBP)-positive oligodendrocytes. Overexpression of DR6 in oligodendrocytes leads to caspase 3 (casp3) activation and cell death. Attenuation of DR6 function leads to enhanced oligodendrocyte maturation, myelination and downregulation of casp3. Treatment with a DR6 antagonist antibody promotes remyelination in both lysolecithin-induced demyelination and experimental autoimmune encephalomyelitis (EAE) models. Consistent with the DR6 antagoinst antibody studies, DR6-null mice show enhanced remyelination in both demyelination models. These studies reveal a pivotal role for DR6 signaling in immature oligodendrocyte maturation and myelination that may provide new therapeutic avenues for the treatment of demyelination disorders such as multiple sclerosis.
Collapse
Affiliation(s)
- Sha Mi
- Biogen Idec, Cambridge, Massachusetts, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Hansen-Pupp I, Hövel H, Hellström A, Hellström-Westas L, Löfqvist C, Larsson EM, Lazeyras F, Fellman V, Hüppi PS, Ley D. Postnatal decrease in circulating insulin-like growth factor-I and low brain volumes in very preterm infants. J Clin Endocrinol Metab 2011; 96:1129-35. [PMID: 21289247 DOI: 10.1210/jc.2010-2440] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT IGF-I and IGF binding protein-3 (IGFBP-3) are essential for growth and maturation of the developing brain. OBJECTIVE The aim of this study was to evaluate the association between postnatal serum concentrations of IGF-I and IGFBP-3 and brain volumes at term in very preterm infants. DESIGN Fifty-one infants with a mean (sd) gestational age (GA) of 26.4 (1.9) wk and birth weight (BW) of 888 (288) g were studied, with weekly blood sampling of IGF-I and IGFBP-3 from birth until 35 gestational weeks (GW) and daily calculation of protein and caloric intake. Magnetic resonance images obtained at 40 GW were segmented into total brain, cerebellar, cerebrospinal fluid, gray matter, and unmyelinated white matter volumes. MAIN OUTCOME MEASURES We evaluated brain growth by measuring brain volumes using magnetic resonance imaging. RESULTS Mean IGF-I concentrations from birth to 35 GW correlated with total brain volume, unmyelinated white matter volume, gray matter volume, and cerebellar volume [r = 0.55 (P < 0.001); r = 0.55 (P < 0.001); r = 0.44 (P = 0.002); and r = 0.58 (P < 0.001), respectively]. Similar correlations were observed for IGFBP-3 concentrations. Correlations remained after adjustment for GA, mean protein and caloric intakes, gender, severe brain damage, and steroid treatment. Protein and caloric intakes were not related to brain volumes. Infants with BW small for GA had lower mean concentrations of IGF-I (P = 0.006) and smaller brain volumes (P = 0.001-0.013) than infants with BW appropriate for GA. CONCLUSION Postnatal IGF-I and IGFBP-3 concentrations are positively associated with brain volumes at 40 GW in very preterm infants. Normalization of the IGF-I axis, directly or indirectly, may support normal brain development in very preterm infants.
Collapse
Affiliation(s)
- Ingrid Hansen-Pupp
- Division of Pediatrics, Department of Clinical Sciences, Lund, Lund University Hospital, 221 85 Lund, Sweden.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Kendall GS, Hristova M, Hirstova M, Horn S, Dafou D, Acosta-Saltos A, Almolda B, Zbarsky V, Rumajogee P, Heuer H, Castellano B, Pfeffer K, Nedospasov SA, Peebles DM, Raivich G. TNF gene cluster deletion abolishes lipopolysaccharide-mediated sensitization of the neonatal brain to hypoxic ischemic insult. J Transl Med 2011; 91:328-41. [PMID: 21135813 DOI: 10.1038/labinvest.2010.192] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
In the current study, we explored the role of TNF cluster cytokines on the lipopolysaccharide (LPS)-mediated, synergistic increase in brain injury after hypoxic ischemic insult in postnatal day 7 mice. Pretreatment with moderate doses of LPS (0.3 μg/g) resulted in particularly pronounced synergistic injury within 12 h. Systemic application of LPS alone resulted in a strong upregulation of inflammation-associated cytokines TNFα, LTβ, interleukin (IL) 1β, IL6, chemokines, such as CXCL1, and adhesion molecules E-Selectin, P-Selectin and intercellular adhesion molecule-1 (ICAM1), as well as a trend toward increased LTα levels in day 7 mouse forebrain. In addition, it was also associated with strong activation of brain blood vessel endothelia and local microglial cells. Here, deletion of the entire TNF gene cluster, removing TNFα, LTβ and LTα completely abolished endotoxin-mediated increase in the volume of cerebral infarct. Interestingly, the same deletion also prevented endothelial and microglial activation following application of LPS alone, suggesting the involvement of these cell types in bringing about the LPS-mediated sensitization to neonatal brain injury.
Collapse
Affiliation(s)
- Giles S Kendall
- Perinatal Brain Repair Group, Centre for Perinatal Brain Protection and Repair, Institute of Women's Health, University College London, London, UK.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Reimunde P, Rodicio C, López N, Alonso A, Devesa P, Devesa J. Effects of recombinant growth hormone replacement and physical rehabilitation in recovery of gross motor function in children with cerebral palsy. Ther Clin Risk Manag 2010; 6:585-92. [PMID: 21151628 PMCID: PMC2999511 DOI: 10.2147/tcrm.s14919] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Cerebral palsy is an important health issue that has a strong socioeconomic impact. There is no cure for cerebral palsy, and therapeutic approaches only report small benefits for affected people. In this study we assessed the effects of growth hormone treatment (0.3 μg/kg/day) combined with physical rehabilitation in the recovery of gross motor function in children with growth hormone deficiency and cerebral palsy (four males and six females, mean age 5.63 ± 2.32 years) as compared with that observed in a similar population of cerebral palsy children (five males, five females, mean age 5.9 ± 2.18 years) without growth hormone deficiency treated only with physical rehabilitation for two months. The Gross Motor Function Measure (GMFM-88) and Modified Ashworth Scale were performed before commencing the treatment and after completion thereof. In children with cerebral palsy and growth hormone deficiency, Dimension A (P < 0.02), dimension B (P < 0.02), and dimension C (P < 0.02) of the GMFM-88, and the total score of the test (P < 0.01) significantly improved after the treatment; dimension D and dimension E did not increase, and four of five spastic patients showed a reduction in spasticity. However, in children with cerebral palsy and without growth hormone deficiency, only the total score of the test improved significantly after the treatment period. This indicates that growth hormone replacement therapy was responsible for the large differences observed between both groups in response to physical rehabilitation. We propose that the combined therapy involving growth hormone administration and physical rehabilitation may be a useful therapeutic approach in the recovery of gross motor function in children with growth hormone deficiency and cerebral palsy.
Collapse
Affiliation(s)
- Pedro Reimunde
- Medical Center "Proyecto Foltra", Cacheiras (Teo), A Coruña, Spain
| | | | | | | | | | | |
Collapse
|
48
|
Reimunde P, Quintana A, Castañón B, Casteleiro N, Vilarnovo Z, Otero A, Devesa A, Otero-Cepeda XL, Devesa J. Effects of growth hormone (GH) replacement and cognitive rehabilitation in patients with cognitive disorders after traumatic brain injury. Brain Inj 2010; 25:65-73. [PMID: 21117918 DOI: 10.3109/02699052.2010.536196] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE To assess the effects of growth hormone (GH) treatment combined with cognitive rehabilitation in patients with adult growth hormone deficiency (GHD) and cognitive disorders occurring after traumatic brain injury (TBI). PARTICIPANTS Nineteen adult patients with TBI: GHD was found in 11 of them. INTERVENTION Patients were treated with GH (GHD; sc; 1 mg/day) or vehicle (controls; sc; 1 mg/day); daily cognitive rehabilitation therapy was performed in both groups for 3 months. MAIN OUTCOME MEASURES The GHRH-arginine test established GHD. The neuropsychological test WAIS was performed before commencing the treatment and 3 months after commencing it. RESULTS Controls achieved significant improvements in digits and in manipulative intelligence quotient (IQ) (p < 0.05 vs. baseline). GHD achieved significant improvements in more cognitive parameters: understanding, digits, numbers and incomplete figures (p < 0.05 vs. baseline) and similarities, vocabulary, verbal IQ, manipulative IQ and total IQ (p < 0.01). GHD reached significantly greater improvements than controls in similarities (p < 0.01) and in vocabulary, verbal IQ and total IQ (p < 0.05). CONCLUSION GH administration significantly improved cognitive rehabilitation in GHD patients. Since at the end of treatment period plasma IGF-I levels were similar in both groups it is likely that exogenous GH administration is responsible for the significant differences found.
Collapse
Affiliation(s)
- P Reimunde
- Medical Center Proyecto Foltra, Cacheiras (Teo), A Coruña, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Petcu EB, Smith RA, Miroiu RI, Opris MM. Angiogenesis in old-aged subjects after ischemic stroke: a cautionary note for investigators. JOURNAL OF ANGIOGENESIS RESEARCH 2010; 2:26. [PMID: 21110846 PMCID: PMC3000373 DOI: 10.1186/2040-2384-2-26] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2010] [Accepted: 11/26/2010] [Indexed: 12/12/2022]
Abstract
Angiogenesis represents a form of neovascularisation of exceptional importance in numerous pathological conditions including stroke. In this context it is directly related to neuroregeneration which is seen in close proximity. However, numerous experimental data have been drawn from studies that have ignored the age criterion. This is extremely important as angiogenesis is different in young versus old subjects. Extrapolating data obtained from studies performed in young subjects or "in vitro" to old-age patients could lead to inexact conclusions since the dynamics of angiogenesis is age-dependent. The current review covers the key features of brain senescence including morphological and functional changes related to the brain parenchyma, its vascular network and blood flow which could possibly influence the process of angiogenesis. This is followed by a description of post-stroke angiogenesis and its relationship to neuroregeneration and its modulation by vascular endothelial growth factor (VEGF) and insulin-like growth factor 1 (IGF 1), the most important factors active in old brain after ischemic injury.
Collapse
Affiliation(s)
- Eugen B Petcu
- Griffith University School of Medicine, Gold Coast Campus, Griffith University, QLD 4222, Australia.
| | | | | | | |
Collapse
|
50
|
Guan J. Insulin-like growth factor -1 (IGF-1) derived neuropeptides, a novel strategy for the development of pharmaceuticals for managing ischemic brain injury. CNS Neurosci Ther 2010; 17:250-5. [PMID: 20236140 DOI: 10.1111/j.1755-5949.2009.00128.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Insulin-Like Growth Factor-1 (IGF-1) is neuroprotective and improves long-term function after brain injury. However, its clinical application to neurological disorders is limited by its large molecular size, poor central uptake, and mitogenic potential. Glycine-proline-glutamate (GPE) is naturally cleaved from the IGF-1 N-terminal and is also neuroprotective after ischemic injury, thus providing a potential novel strategy of drug discovery for management of neurological disorders. GPE is not enzymatically stable, thus intravenous infusion of GPE becomes necessary for stable and potent neuroprotection. The broad effective dose range and treatment window of 3-7 h after the lesion suggest its potential for treating acute brain injuries. The neuroprotective action of GPE is not age selective, is not dependent on cerebral reperfusion, plasma glucose concentrations, and core body temperature. G-2mPE, a GPE analogue designed to be more resistant to enzymatic activity, has a prolonged plasma half-life and is more potent in neuroprotection. Neuroprotection by GPE and its analogue may be involved in modulation of inflammation, promotion of astrocytosis, inhibition of apoptosis, and in vascular remodeling. Small neuropeptides have advantages over growth factors in the treatment of brain injury, and modified neuropeptides, designed to overcome the limitations of their endogenous counterparts, represent a novel strategy of pharmaceutical discovery for neurological disorders.
Collapse
Affiliation(s)
- Jian Guan
- Liggins Institute, The University of Auckland, New Zealand.
| |
Collapse
|