1
|
Cai Y, Wang T. Regulation of presynaptic homeostatic plasticity by glial signalling in Alzheimer's disease. J Physiol 2024. [PMID: 39705214 DOI: 10.1113/jp286751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 12/04/2024] [Indexed: 12/22/2024] Open
Abstract
Alzheimer's disease (AD), the most common form of dementia among the elderly, affects numerous individuals worldwide. Despite advances in understanding the molecular underpinnings of AD pathology, effective treatments to prevent or cure the disease remain elusive. AD is characterized not only by pathological hallmarks such as amyloid plaques and neurofibrillary tangles but also by impairments in synaptic physiology, circuit activity and cognitive function. Synaptic homeostatic plasticity plays a vital role in maintaining the stability of synaptic and neural functions amid genetic and environmental disturbances. A key component of this regulation is presynaptic homeostatic potentiation, where increased presynaptic neurotransmitter release compensates for reduced postsynaptic glutamate receptor functionality, thereby stabilizing neuronal excitability. The role of presynaptic homeostatic plasticity in synapse stabilization in AD, however, remains unclear. Moreover, recent advances in transcriptomics have illuminated the complex roles of glial cells in regulating synaptic function in ageing brains and in the progression of neurodegenerative diseases. Yet, the impact of AD-related abnormalities in glial signalling on synaptic homeostatic plasticity has not been fully delineated. This review discusses recent findings on how glial dysregulation in AD affects presynaptic homeostatic plasticity. There is increasing evidence that disrupted glial signalling, particularly through aberrant histone acetylation and transcriptomic changes in glia, compromises this plasticity in AD. Notably, the sphingosine signalling pathway has been identified as being protective in stabilizing synaptic physiology through epigenetic and homeostatic mechanisms, presenting potential therapeutic targets for treating neurodegenerative disorders.
Collapse
Affiliation(s)
- Yimei Cai
- Department of Pharmacology & Physiology, Georgetown University Medical Center, Washington, D.C., USA
| | - Tingting Wang
- Department of Pharmacology & Physiology, Georgetown University Medical Center, Washington, D.C., USA
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, D.C., USA
| |
Collapse
|
2
|
Huré JB, Foucault L, Ghayad LM, Marie C, Vachoud N, Baudouin L, Azmani R, Ivljanin N, Arevalo-Nuevo A, Pigache M, Bouslama-Oueghlani L, Chemelle JA, Dronne MA, Terreux R, Hassan B, Gueyffier F, Raineteau O, Parras C. Pharmacogenomic screening identifies and repurposes leucovorin and dyclonine as pro-oligodendrogenic compounds in brain repair. Nat Commun 2024; 15:9837. [PMID: 39537633 PMCID: PMC11561360 DOI: 10.1038/s41467-024-54003-9] [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: 05/03/2023] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
Oligodendrocytes are critical for CNS myelin formation and are involved in preterm-birth brain injury (PBI) and multiple sclerosis (MS), both of which lack effective treatments. We present a pharmacogenomic approach that identifies compounds with potent pro-oligodendrogenic activity, selected through a scoring strategy (OligoScore) based on their modulation of oligodendrogenic and (re)myelination-related transcriptional programs. Through in vitro neural and oligodendrocyte progenitor cell (OPC) cultures, ex vivo cerebellar explants, and in vivo mouse models of PBI and MS, we identify FDA-approved leucovorin and dyclonine as promising candidates. In a neonatal chronic hypoxia mouse model mimicking PBI, both compounds promote neural progenitor cell proliferation and oligodendroglial fate acquisition, with leucovorin further enhancing differentiation. In an adult MS model of focal de/remyelination, they improve lesion repair by promoting OPC differentiation while preserving the OPC pool. Additionally, they shift microglia from a pro-inflammatory to a pro-regenerative profile and enhance myelin debris clearance. These findings support the repurposing of leucovorin and dyclonine for clinical trials targeting myelin disorders, offering potential therapeutic avenues for PBI and MS.
Collapse
Affiliation(s)
- Jean-Baptiste Huré
- Paris Brain Institute, Sorbonne Université, Inserm U1127, CNRS UMR 7225, Hôpital Pitié-Salpêtrière, Paris, France
| | - Louis Foucault
- Univ Lyon, Université Claude Bernard Lyon1, Inserm, Stem Cell and Brain Research Institute U1208, Bron, France
| | - Litsa Maria Ghayad
- Paris Brain Institute, Sorbonne Université, Inserm U1127, CNRS UMR 7225, Hôpital Pitié-Salpêtrière, Paris, France
| | - Corentine Marie
- Paris Brain Institute, Sorbonne Université, Inserm U1127, CNRS UMR 7225, Hôpital Pitié-Salpêtrière, Paris, France
| | - Nicolas Vachoud
- Univ Lyon, Université Claude Bernard Lyon1, Inserm, Stem Cell and Brain Research Institute U1208, Bron, France
| | - Lucas Baudouin
- Paris Brain Institute, Sorbonne Université, Inserm U1127, CNRS UMR 7225, Hôpital Pitié-Salpêtrière, Paris, France
| | - Rihab Azmani
- Univ Lyon, Université Claude Bernard Lyon1, Inserm, Stem Cell and Brain Research Institute U1208, Bron, France
| | - Natalija Ivljanin
- Paris Brain Institute, Sorbonne Université, Inserm U1127, CNRS UMR 7225, Hôpital Pitié-Salpêtrière, Paris, France
| | - Alvaro Arevalo-Nuevo
- Paris Brain Institute, Sorbonne Université, Inserm U1127, CNRS UMR 7225, Hôpital Pitié-Salpêtrière, Paris, France
| | - Morgane Pigache
- Paris Brain Institute, Sorbonne Université, Inserm U1127, CNRS UMR 7225, Hôpital Pitié-Salpêtrière, Paris, France
| | - Lamia Bouslama-Oueghlani
- Paris Brain Institute, Sorbonne Université, Inserm U1127, CNRS UMR 7225, Hôpital Pitié-Salpêtrière, Paris, France
| | - Julie-Anne Chemelle
- Équipe ECMO, Laboratoire de Biologie Tissulaire et d'Ingénierie (LBTI), UMR5305, Lyon, France
| | - Marie-Aimée Dronne
- Claude Bernard University, UMR5558 Laboratoire de Biométrie et Biologie Evolutive, CNRS, Villeurbanne, France
| | - Raphaël Terreux
- Équipe ECMO, Laboratoire de Biologie Tissulaire et d'Ingénierie (LBTI), UMR5305, Lyon, France
| | - Bassem Hassan
- Paris Brain Institute, Sorbonne Université, Inserm U1127, CNRS UMR 7225, Hôpital Pitié-Salpêtrière, Paris, France
| | - François Gueyffier
- Claude Bernard University, UMR5558 Laboratoire de Biométrie et Biologie Evolutive, CNRS, Villeurbanne, France
| | - Olivier Raineteau
- Univ Lyon, Université Claude Bernard Lyon1, Inserm, Stem Cell and Brain Research Institute U1208, Bron, France.
| | - Carlos Parras
- Paris Brain Institute, Sorbonne Université, Inserm U1127, CNRS UMR 7225, Hôpital Pitié-Salpêtrière, Paris, France.
| |
Collapse
|
3
|
Omura S, Ogawa R, Kawachi T, Ogawa A, Arai Y, Takayama N, Masui A, Kondo K, Sugimoto H, Shinohara HM, Takahashi T, Maeda H, Ohyama K. Olig2+/NG2+/BLBP+ astrocyte progenitors: a novel component of the neurovascular unit in the developing mouse hippocampus. Front Cell Neurosci 2024; 18:1464402. [PMID: 39484182 PMCID: PMC11524929 DOI: 10.3389/fncel.2024.1464402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 09/30/2024] [Indexed: 11/03/2024] Open
Abstract
Astrocytes are key components of the neurovascular unit. While we have recently identified Olig2+ astrocyte progenitors (ASPs) in the developing mouse dentate gyrus (DG), their molecular signature remains incompletely characterized. Here we demonstrate that Olig2+ ASPs predominantly express brain lipid-binding protein (BLBP), while only a small population of them expresses gfap-GFP. These Olig2+/BLBP+ ASPs co-express the transcription factors Sox3, Sox9 and the proteoglycan NG2 but not Sox10, a marker for oligodendrocyte progenitors (OLPs). Olig2+ ASPs appear from embryonic day 18 (E18) onwards and decline at postnatal day 14 (P14). Consistent with the proliferation of both Olig2+ and NG2+ glial cells after brain injury, intrauterine intermittent hypoxia (IH) led to an increase in Olig2+/NG2+/BLBP+ ASPs in the postnatal DG. IH also promoted both angiogenesis and vascular coupling of Olig2+/NG2+ ASPs. Our data suggest that IH-induced expression of HIF1a increases Olig2+/NG2+/BLBP+ ASPs in a cell non-autonomous manner. Our data also revealed increased vascular coupling of GFAP+ astrocytes following IH, while the number of GFAP+ astrocytes remains unchanged. Given that BLBP, Olig2 and NG2 are expressed in reactive astrocytes, our findings suggest that Olig2+/NG2+/BLBP+ ASPs represent a subtype of reactive astrocyte progenitors. Furthermore, the enhanced vascular coupling of Olig2+/NG2+/BLBP+ ASPs appears to be an adaptive response to hypoxic brain injury. This study provides new insights into the molecular characteristics of Olig2+/NG2+/BLBP+ ASPs and their potential role in the brain's response to hypoxic injury, contributing to our understanding of neurovascular unit dynamics in both development and pathological conditions.
Collapse
Affiliation(s)
- Shoichiro Omura
- Department of Histology and Neuroanatomy, Tokyo Medical University, Tokyo, Japan
| | - Rina Ogawa
- Department of Histology and Neuroanatomy, Tokyo Medical University, Tokyo, Japan
| | - Tomomi Kawachi
- Department of Histology and Neuroanatomy, Tokyo Medical University, Tokyo, Japan
| | - Aya Ogawa
- Department of Histology and Neuroanatomy, Tokyo Medical University, Tokyo, Japan
| | - Yuuki Arai
- Department of Histology and Neuroanatomy, Tokyo Medical University, Tokyo, Japan
| | - Natsumi Takayama
- Department of Histology and Neuroanatomy, Tokyo Medical University, Tokyo, Japan
| | - Aki Masui
- Department of Histology and Neuroanatomy, Tokyo Medical University, Tokyo, Japan
| | - Kumiko Kondo
- Department of Histology and Neuroanatomy, Tokyo Medical University, Tokyo, Japan
| | - Hiroki Sugimoto
- Department of Histology and Neuroanatomy, Tokyo Medical University, Tokyo, Japan
| | - Hiroshi M. Shinohara
- Department of Histology and Neuroanatomy, Tokyo Medical University, Tokyo, Japan
| | - Tokiharu Takahashi
- Department of Histology and Neuroanatomy, Tokyo Medical University, Tokyo, Japan
| | - Hideyuki Maeda
- Department of Legal Medicine, Osaka University, Suita, Japan
| | - Kyoji Ohyama
- Department of Histology and Neuroanatomy, Tokyo Medical University, Tokyo, Japan
| |
Collapse
|
4
|
Odell EP, Jabassini N, Schniedewind B, Pease-Raissi SE, Frymoyer A, Christians U, Green AJ, Chan JR, Ostrem BEL. Minimum effective dose of clemastine in a mouse model of preterm white matter injury. Pediatr Res 2024; 96:933-941. [PMID: 38942888 PMCID: PMC11502491 DOI: 10.1038/s41390-024-03326-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 05/06/2024] [Accepted: 05/31/2024] [Indexed: 06/30/2024]
Abstract
BACKGROUND Preterm white matter injury (PWMI) is the most common cause of brain injury in premature neonates. PWMI involves a differentiation arrest of oligodendrocytes, the myelinating cells of the central nervous system. Clemastine was previously shown to induce oligodendrocyte differentiation and myelination in mouse models of PWMI at a dose of 10 mg/kg/day. The minimum effective dose (MED) of clemastine is unknown. Identification of the MED is essential for maximizing safety and efficacy in neonatal clinical trials. We hypothesized that the MED in neonatal mice is lower than 10 mg/kg/day. METHODS Mouse pups were exposed to normoxia or hypoxia (10% FiO2) from postnatal day 3 (P3) through P10. Vehicle or clemastine at one of four doses (0.5, 2, 7.5 or 10 mg/kg/day) was given to hypoxia-exposed pups. Myelination was assessed at age P14 and 10 weeks to determine the MED. Clemastine pharmacokinetics were evaluated at steady-state on day 8 of treatment. RESULTS Clemastine rescued hypoxia-induced hypomyelination with a MED of 7.5 mg/kg/day. Pharmacokinetic analysis of the MED revealed Cmax 44.0 ng/mL, t1/2 4.6 h, and AUC24 280.1 ng*hr/mL. CONCLUSIONS Based on these results, myelination-promoting exposures should be achievable with oral doses of clemastine in neonates with PWMI. IMPACT Preterm white matter injury (PWMI) is the most common cause of brain injury and cerebral palsy in premature neonates. Clemastine, an FDA-approved antihistamine, was recently identified to strongly promote myelination in a mouse model of PWMI and is a possible treatment. The minimum effective dose in neonatal rodents is unknown and is critical for guiding dose selection and balancing efficacy with toxicity in future clinical trials. We identified the minimum effective dose of clemastine and the associated pharmacokinetics in a murine chronic hypoxia model of PWMI, paving the way for a future clinical trial in human neonates.
Collapse
Affiliation(s)
- Elizabeth P Odell
- Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Nora Jabassini
- Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Björn Schniedewind
- iC42 Clinical Research and Development, Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Sarah E Pease-Raissi
- Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Adam Frymoyer
- Department of Pediatrics, Stanford University, Palo Alto, CA, USA
| | - Uwe Christians
- iC42 Clinical Research and Development, Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Ari J Green
- Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, USA
| | - Jonah R Chan
- Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Bridget E L Ostrem
- Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
5
|
Feng Y, Huang Z, Ma X, Zong X, Xu P, Lin HW, Zhang Q. Intermittent theta-burst stimulation alleviates hypoxia-ischemia-caused myelin damage and neurologic disability. Exp Neurol 2024; 378:114821. [PMID: 38782349 PMCID: PMC11214828 DOI: 10.1016/j.expneurol.2024.114821] [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: 12/04/2023] [Revised: 05/01/2024] [Accepted: 05/19/2024] [Indexed: 05/25/2024]
Abstract
Neonatal hypoxia-ischemia (HI) results in behavioral deficits, characterized by neuronal injury and retarded myelin formation. To date, limited treatment methods are available to prevent or alleviate neurologic sequelae of HI. Intermittent theta-burst stimulation (iTBS), a non-invasive therapeutic procedure, is considered a promising therapeutic tool for treating some neurocognitive disorders and neuropsychiatric diseases. Hence, this study aims to investigate whether iTBS can prevent the negative behavioral manifestations of HI and explore the mechanisms for associations. We exposed postnatal day 10 Sprague-Dawley male and female rats to 2 h of hypoxia (6% O2) following right common carotid artery ligation, resulting in oligodendrocyte (OL) dysfunction, including reduced proliferation and differentiation of oligodendrocyte precursor cells (OPCs), decreased OL survival, and compromised myelin in the corpus callosum (CC) and hippocampal dentate gyrus (DG). These alterations were concomitant with cognitive dysfunction and depression-like behaviors. Crucially, early iTBS treatment (15 G, 190 s, seven days, initiated one day post-HI) significantly alleviated HI-caused myelin damage and mitigated the neurologic sequelae both in male and female rats. However, the late iTBS treatment (initiated 18 days after HI insult) could not significantly impact these behavioral deficits. In summary, our findings support that early iTBS treatment may be a promising strategy to improve HI-induced neurologic disability. The underlying mechanisms of iTBS treatment are associated with promoting the differentiation of OPCs and alleviating myelin damage.
Collapse
Affiliation(s)
- Yu Feng
- Department of Neurology, Louisiana State University Health Sciences Center, Shreveport, 1501 Kings Highway, LA 71103, USA
| | - Zhihai Huang
- Department of Neurology, Louisiana State University Health Sciences Center, Shreveport, 1501 Kings Highway, LA 71103, USA
| | - Xiaohui Ma
- Department of Neurology, Louisiana State University Health Sciences Center, Shreveport, 1501 Kings Highway, LA 71103, USA
| | - Xuemei Zong
- Department of Neurology, Louisiana State University Health Sciences Center, Shreveport, 1501 Kings Highway, LA 71103, USA
| | - Peisheng Xu
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina, College of Pharmacy, 715 Sumter Street, CLS609D, Columbia, SC 29208, USA
| | - Hung Wen Lin
- Department of Neurology, Louisiana State University Health Sciences Center, Shreveport, 1501 Kings Highway, LA 71103, USA
| | - Quanguang Zhang
- Department of Neurology, Louisiana State University Health Sciences Center, Shreveport, 1501 Kings Highway, LA 71103, USA.
| |
Collapse
|
6
|
Odell E, Jabassini N, Schniedewind B, Pease-Raissi SE, Frymoyer A, Christians U, Green AJ, Chan JR, Ostrem BEL. Minimum Effective Dose of Clemastine in a Mouse Model of Preterm White Matter Injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.08.578953. [PMID: 38464078 PMCID: PMC10925142 DOI: 10.1101/2024.02.08.578953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Background Preterm white matter injury (PWMI) is the most common cause of brain injury in premature neonates. PWMI involves a differentiation arrest of oligodendrocytes, the myelinating cells of the central nervous system. Clemastine was previously shown to induce oligodendrocyte differentiation and myelination in mouse models of PWMI at a dose of 10 mg/kg/day. The minimum effective dose (MED) of clemastine is unknown. Identification if the MED is essential for maximizing safety and efficacy in neonatal clinical trials. We hypothesized that the MED in neonatal mice is lower than 10 mg/kg/day. Methods Mouse pups were exposed to normoxia or hypoxia (10% FiO 2 ) from postnatal day 3 (P3) through P10. Vehicle or clemastine fumarate at one of four doses (0.5, 2, 7.5 or 10 mg/kg/day) was given orally to hypoxia-exposed pups. At P14, myelination was assessed by immunohistochemistry and electron microscopy to determine the MED. Clemastine pharmacokinetics were evaluated at steady-state on day 8 of treatment. Results Clemastine rescued hypoxia-induced hypomyelination with a MED of 7.5 mg/kg/day. Pharmacokinetic analysis of the MED revealed C max 44.0 ng/mL, t 1/2 4.6 hours, and AUC 24 280.1 ng*hr/mL. Conclusion Based on these results, myelination-promoting exposures should be achievable with oral doses of clemastine in neonates with PWMI. Key Points Preterm white matter injury (PWMI) is the most common cause of brain injury and cerebral palsy in premature neonates.Clemastine, an FDA-approved antihistamine, was recently identified to strongly promote myelination in a mouse model of PWMI and is a possible treatment.The minimum effective dose in neonatal rodents is unknown and is critical for guiding dose selection and balancing efficacy with toxicity in future clinical trials.We identified the minimum effective dose of clemastine and the associated pharmacokinetics in a murine chronic hypoxia model of PWMI, paving the way for a future clinical trial in human neonates.
Collapse
|
7
|
Van Steenwinckel J, Bokobza C, Laforge M, Shearer IK, Miron VE, Rua R, Matta SM, Hill‐Yardin EL, Fleiss B, Gressens P. Key roles of glial cells in the encephalopathy of prematurity. Glia 2024; 72:475-503. [PMID: 37909340 PMCID: PMC10952406 DOI: 10.1002/glia.24474] [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: 07/19/2023] [Revised: 09/17/2023] [Accepted: 09/19/2023] [Indexed: 11/03/2023]
Abstract
Across the globe, approximately one in 10 babies are born preterm, that is, before 37 weeks of a typical 40 weeks of gestation. Up to 50% of preterm born infants develop brain injury, encephalopathy of prematurity (EoP), that substantially increases their risk for developing lifelong defects in motor skills and domains of learning, memory, emotional regulation, and cognition. We are still severely limited in our abilities to prevent or predict preterm birth. No longer just the "support cells," we now clearly understand that during development glia are key for building a healthy brain. Glial dysfunction is a hallmark of EoP, notably, microgliosis, astrogliosis, and oligodendrocyte injury. Our knowledge of glial biology during development is exponentially expanding but hasn't developed sufficiently for development of effective neuroregenerative therapies. This review summarizes the current state of knowledge for the roles of glia in infants with EoP and its animal models, and a description of known glial-cell interactions in the context of EoP, such as the roles for border-associated macrophages. The field of perinatal medicine is relatively small but has worked passionately to improve our understanding of the etiology of EoP coupled with detailed mechanistic studies of pre-clinical and human cohorts. A primary finding from this review is that expanding our collaborations with computational biologists, working together to understand the complexity of glial subtypes, glial maturation, and the impacts of EoP in the short and long term will be key to the design of therapies that improve outcomes.
Collapse
Affiliation(s)
| | - Cindy Bokobza
- NeuroDiderot, INSERMUniversité Paris CitéParisFrance
| | | | - Isabelle K. Shearer
- School of Health and Biomedical SciencesSTEM College, RMIT UniversityBundooraVictoriaAustralia
| | - Veronique E. Miron
- Barlo Multiple Sclerosis CentreSt. Michael's HospitalTorontoOntarioCanada
- Department of ImmunologyUniversity of TorontoTorontoOntarioCanada
- College of Medicine and Veterinary MedicineThe Dementia Research Institute at The University of EdinburghEdinburghUK
| | - Rejane Rua
- CNRS, INSERM, Centre d'Immunologie de Marseille‐Luminy (CIML), Turing Centre for Living SystemsAix‐Marseille UniversityMarseilleFrance
| | - Samantha M. Matta
- School of Health and Biomedical SciencesSTEM College, RMIT UniversityBundooraVictoriaAustralia
| | - Elisa L. Hill‐Yardin
- School of Health and Biomedical SciencesSTEM College, RMIT UniversityBundooraVictoriaAustralia
| | - Bobbi Fleiss
- NeuroDiderot, INSERMUniversité Paris CitéParisFrance
- School of Health and Biomedical SciencesSTEM College, RMIT UniversityBundooraVictoriaAustralia
| | | |
Collapse
|
8
|
Lin Q, Lin L, Li L, Zheng YF, Hu DW, Zhang G. Dynamic changes of oligodendrogenesis in neonatal rats with hypoxic-ischemic white matter injury. Brain Res 2023; 1817:148495. [PMID: 37481153 DOI: 10.1016/j.brainres.2023.148495] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 07/18/2023] [Accepted: 07/19/2023] [Indexed: 07/24/2023]
Abstract
BACKGROUND White matter injury (WMI) is an important type of preterm brain injury, which may result in severe neurological sequelae and lack of effective treatments. It is ascertained that selective vulnerability of oligodendrocytes is closely related to the WMI in preterm infants. But the alteration of the endogenous oligodendrogenesis over long time after hypoxic-ischemic WMI is still not clearly elucidated. METHODS We adopted an animal model of hypoxic-ischemic WMI in 3-day-old neonatal Sprague-Dawley rats. Immunofluorescence staining and western blotting were used to detect dynamic changes of oligodendrogenesis in the white matter region on postoperative day (POD) 1, 3, 7, 14, 28, 56 and 84. RESULTS In the sham group, the oligodendrocyte lineage in the white matter reached a developmental peak from POD 3 to 14. The proliferation and development of oligodendrocyte precursor cells (OPCs) occurred primarily within POD 14. The number of mature oligodendrocytes showed an upward trend and a dynamic change in proliferation over time. While in the WMI group, the oligodendrocyte lineage was upregulated on POD1 and 3 but downregulated on POD 7 and 14. The proliferation of OPCs increased on POD 1 and decreased on POD 3 and 7, with the total number of OPCs significantly reduced from POD 3 to 14. The number of mature oligodendrocytes decreased from POD 3 to 28, and return to the level of the sham group on POD 56 and 84, whereas the MBP expression was still significantly downregulated on POD 56 and 84. CONCLUSIONS Hypoxia-ischemia can have a long-term dynamic effect on the endogenous oligodendrogenesis of neonatal rat brain white matter. The proliferation of OPCs was promoted on POD 1 but inhibited from POD 3 to 14, which may be an early intervention target to improve oligodendrogenesis. The number of mature oligodendrocytes recover to the normal on POD 56 and 84 but the myelination is still blocked, which suggests it is essential to promote the maturation of oligodendrocyte and its function recovery at the same time within POD 28. Such efforts will provide the opportunity to test new interventions in pre-clinical studies for their promising clinical application.
Collapse
Affiliation(s)
- Qing Lin
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative Diseases, Fujian Medical University, Fuzhou, China; Laboratory of Clinical Applied Anatomy, Department of Human Anatomy, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Ling Lin
- Public Technology Service Center, Fujian Medical University, Fuzhou, China
| | - Li Li
- Assisted Reproduction Centre, Obstetrics and Gynecology Department, 900TH Hospital of Joint Logistics Support Force, Fuzhou, China
| | - Yu-Fen Zheng
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative Diseases, Fujian Medical University, Fuzhou, China
| | - Ding-Wang Hu
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative Diseases, Fujian Medical University, Fuzhou, China; Laboratory of Clinical Applied Anatomy, Department of Human Anatomy, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China.
| | - Geng Zhang
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative Diseases, Fujian Medical University, Fuzhou, China; Laboratory of Clinical Applied Anatomy, Department of Human Anatomy, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
9
|
Xie Q, Hua X, Huang C, Liao X, Tian Z, Xu J, Zhao Y, Jiang G, Huang H, Huang C. SOX2 Promotes Invasion in Human Bladder Cancers through MMP2 Upregulation and FOXO1 Downregulation. Int J Mol Sci 2022; 23:ijms232012532. [PMID: 36293387 PMCID: PMC9604292 DOI: 10.3390/ijms232012532] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/03/2022] [Accepted: 10/11/2022] [Indexed: 11/16/2022] Open
Abstract
SOX2, a member of the SRY-related HMG-box (SOX) family, is abnormally expressed in many tumors and associated with cancer stem cell-like properties. Previous reports have shown that SOX2 is a biomarker for cancer stem cells in human bladder cancer (BC), and our most recent study has indicated that the inhibition of SOX2 by anticancer compound ChlA-F attenuates human BC cell invasion. We now investigated the mechanisms through which SOX2 promotes the invasive ability of BC cells. Our studies revealed that SOX2 promoted SKP2 transcription and increased SKP2-accelerated Sp1 protein degradation. As Sp1 is a transcriptionally regulated gene, HUR transcription was thereby attenuated, and, in the absence of HUR, FOXO1 mRNA was degraded fast, which promoted BC cell invasion. In addition, SOX2 promoted BC invasion through the upregulation of nucleolin transcription, which resulted in increased MMP2 mRNA stability and expression. Collectively, our findings show that SOX2 promotes BC invasion through both SKP2-Sp1-HUR-FOXO1 and nucleolin-MMP2 dual axes.
Collapse
Affiliation(s)
- Qipeng Xie
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325000, China
- Department of Clinical Laboratory, The Second Affiliated Hospital & Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Xiaohui Hua
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325000, China
| | - Chao Huang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Correspondence: (C.H.); (C.H.); Tel.: +86-135-2288-7554 (Chuanshu Huang)
| | - Xin Liao
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zhongxian Tian
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325000, China
| | - Jiheng Xu
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325000, China
| | - Yunping Zhao
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325000, China
| | - Guosong Jiang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Haishan Huang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325000, China
| | - Chuanshu Huang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325000, China
- Correspondence: (C.H.); (C.H.); Tel.: +86-135-2288-7554 (Chuanshu Huang)
| |
Collapse
|
10
|
Jablonska B, Adams KL, Kratimenos P, Li Z, Strickland E, Haydar TF, Kusch K, Nave KA, Gallo V. Sirt2 promotes white matter oligodendrogenesis during development and in models of neonatal hypoxia. Nat Commun 2022; 13:4771. [PMID: 35970992 PMCID: PMC9378658 DOI: 10.1038/s41467-022-32462-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 08/01/2022] [Indexed: 01/02/2023] Open
Abstract
Delayed oligodendrocyte (OL) maturation caused by hypoxia (Hx)-induced neonatal brain injury results in hypomyelination and leads to neurological disabilities. Previously, we characterized Sirt1 as a crucial regulator of OL progenitor cell (OPC) proliferation in response to Hx. We now identify Sirt2 as a critical promoter of OL differentiation during both normal white matter development and in a mouse model of Hx. Importantly, we find that Hx reduces Sirt2 expression in mature OLs and that Sirt2 overexpression in OPCs restores mature OL populations. Reduced numbers of Sirt2+ OLs were also observed in the white matter of preterm human infants. We show that Sirt2 interacts with p27Kip1/FoxO1, p21Cip1/Cdk4, and Cdk5 pathways, and that these interactions are altered by Hx. Furthermore, Hx induces nuclear translocation of Sirt2 in OPCs where it binds several genomic targets. Overall, these results indicate that a balance of Sirt1 and Sirt2 activity is required for developmental oligodendrogenesis, and that these proteins represent potential targets for promoting repair following white matter injury.
Collapse
Affiliation(s)
- Beata Jablonska
- Center for Neuroscience Research, Children's National Research Institute, Children's National Hospital, Washington, DC, 20010, USA.
| | - Katrina L Adams
- Center for Neuroscience Research, Children's National Research Institute, Children's National Hospital, Washington, DC, 20010, USA
| | - Panagiotis Kratimenos
- Center for Neuroscience Research, Children's National Research Institute, Children's National Hospital, Washington, DC, 20010, USA
- Neonatology Department, Children's National Hospital, Washington, DC, 20010, USA
| | - Zhen Li
- Center for Neuroscience Research, Children's National Research Institute, Children's National Hospital, Washington, DC, 20010, USA
| | - Emma Strickland
- Center for Neuroscience Research, Children's National Research Institute, Children's National Hospital, Washington, DC, 20010, USA
| | - Tarik F Haydar
- Center for Neuroscience Research, Children's National Research Institute, Children's National Hospital, Washington, DC, 20010, USA
| | - Katharina Kusch
- Max Planck Institute of Experimental Medicine, Department of Neurogenetics, Gottingen, Germany
| | - Klaus-Armin Nave
- Max Planck Institute of Experimental Medicine, Department of Neurogenetics, Gottingen, Germany
| | - Vittorio Gallo
- Center for Neuroscience Research, Children's National Research Institute, Children's National Hospital, Washington, DC, 20010, USA.
| |
Collapse
|
11
|
Nowacki JC, Fields AM, Fu MM. Emerging cellular themes in leukodystrophies. Front Cell Dev Biol 2022; 10:902261. [PMID: 36003149 PMCID: PMC9393611 DOI: 10.3389/fcell.2022.902261] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 06/30/2022] [Indexed: 11/18/2022] Open
Abstract
Leukodystrophies are a broad spectrum of neurological disorders that are characterized primarily by deficiencies in myelin formation. Clinical manifestations of leukodystrophies usually appear during childhood and common symptoms include lack of motor coordination, difficulty with or loss of ambulation, issues with vision and/or hearing, cognitive decline, regression in speech skills, and even seizures. Many cases of leukodystrophy can be attributed to genetic mutations, but they have diverse inheritance patterns (e.g., autosomal recessive, autosomal dominant, or X-linked) and some arise from de novo mutations. In this review, we provide an updated overview of 35 types of leukodystrophies and focus on cellular mechanisms that may underlie these disorders. We find common themes in specialized functions in oligodendrocytes, which are specialized producers of membranes and myelin lipids. These mechanisms include myelin protein defects, lipid processing and peroxisome dysfunction, transcriptional and translational dysregulation, disruptions in cytoskeletal organization, and cell junction defects. In addition, non-cell-autonomous factors in astrocytes and microglia, such as autoimmune reactivity, and intercellular communication, may also play a role in leukodystrophy onset. We hope that highlighting these themes in cellular dysfunction in leukodystrophies may yield conceptual insights on future therapeutic approaches.
Collapse
|
12
|
Lin WS, Lin SJ, Liao PY, Suresh D, Hsu TR, Wang PY. Role of Ketogenic Diets in Multiple Sclerosis and Related Animal Models: An Updated Review. Adv Nutr 2022; 13:2002-2014. [PMID: 35679067 PMCID: PMC9526852 DOI: 10.1093/advances/nmac065] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 04/25/2022] [Accepted: 06/02/2022] [Indexed: 01/28/2023] Open
Abstract
Prescribing a ketogenic diet (KD) is a century-old dietary intervention mainly used in the context of intractable epilepsy. The classic KD and its variants regained popularity in recent decades, and they are considered potentially beneficial in a variety of neurological conditions other than epilepsy. Many patients with multiple sclerosis (MS) have attempted diet modification for better control of their disease, although evidence thus far remains insufficient to recommend a specific diet for these patients. The results of 3 pilot clinical trials of KD therapy for MS, as well as several related studies, have been reported in recent years. The preliminary findings suggest that KD is safe, feasible, and potentially neuroprotective and disease-modifying for patients with MS. Research on corresponding rodent models has also lent support to the efficacy of KD in the prevention and treatment of experimental autoimmune encephalomyelitis and toxin-induced inflammatory demyelinating conditions in the brain. Furthermore, the animal studies have yielded mechanistic insights into the molecular mechanisms of KD action in relevant situations, paving the way for precision nutrition. Herein we review and synthesize recent advances and also identify unresolved issues, such as the roles of adipokines and gut microbiota, in this field. Hopefully this panoramic view of current understanding can inform future research directions and clinical practice with regard to KD in MS and related conditions.
Collapse
Affiliation(s)
| | - Shan-Ju Lin
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital Yunlin Branch, Yunlin, Taiwan
| | - Pei-Yin Liao
- Department of Dietetics, National Taiwan University Hospital Yunlin Branch, Yunlin, Taiwan
| | - Divya Suresh
- Department of Pediatrics, National Taiwan University Hospital Yunlin Branch, Yunlin, Taiwan
| | - Ting-Rong Hsu
- Department of Pediatrics, Taipei Veterans General Hospital, Taipei, Taiwan,School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Pei-Yu Wang
- Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei, Taiwan,Neurobiology and Cognitive Science Center, National Taiwan University, Taipei, Taiwan,Program in Translational Medicine, National Taiwan University and Academia Sinica, Taipei, Taiwan,Taiwan International Graduate Program in Interdisciplinary Neuroscience, National Taiwan University and Academia Sinica, Taipei, Taiwan,Graduate Institute of Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
13
|
Dion V, Schumacher N, Masar N, Pieltain A, Tocquin P, Lesoinne P, Malgrange B, Vandenbosch R, Franzen R. Cyclin-dependent kinase 7 contributes to myelin maintenance in the adult central nervous system and promotes myelin gene expression. Glia 2022; 70:1652-1665. [PMID: 35488490 DOI: 10.1002/glia.24186] [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: 05/05/2021] [Revised: 04/08/2022] [Accepted: 04/21/2022] [Indexed: 11/08/2022]
Abstract
Mechanisms regulating oligodendrocyte differentiation, developmental myelination and myelin maintenance in adulthood are complex and still not completely described. Their understanding is crucial for the development of new protective or therapeutic strategies in demyelinating pathologies such as multiple sclerosis. In this perspective, we have investigated the role of Cyclin-dependent kinase 7 (Cdk7), a kinase involved in cell-cycle progression and transcription regulation, in the oligodendroglial lineage. We generated a conditional knock-out mouse model in which Cdk7 is invalidated in post-mitotic oligodendrocytes. At the end of developmental myelination, the number and diameter of myelinated axons, as well as the myelin structure, thickness and protein composition, were normal. However, in young adult and in aged mice, there was a higher number of small caliber myelinated axons associated with a decreased mean axonal diameter, myelin sheaths of large caliber axons were thinner, and the level of some major myelin-associated proteins was reduced. These defects were accompanied by the appearance of an abnormal clasping phenotype. We also used an in vitro oligodendroglial model and showed that Cdk7 pharmacological inhibition led to an altered myelination-associated morphological modification combined with a decreased expression of myelin-specific genes. Altogether, we identified novel functions for Cdk7 in CNS myelination.
Collapse
Affiliation(s)
- Valérie Dion
- Laboratory of Nervous System Disorders and Therapies, GIGA Neurosciences, University of Liège, Liège, Belgium
| | - Nathalie Schumacher
- Laboratory of Nervous System Disorders and Therapies, GIGA Neurosciences, University of Liège, Liège, Belgium
| | - Nathalie Masar
- Laboratory of Nervous System Disorders and Therapies, GIGA Neurosciences, University of Liège, Liège, Belgium
| | - Alexandra Pieltain
- Laboratory of Nervous System Disorders and Therapies, GIGA Neurosciences, University of Liège, Liège, Belgium
| | - Pierre Tocquin
- CARE PhytoSYSTEMS, Integrative Biological Sciences, University of Liège, Liège, Belgium
| | - Pierre Lesoinne
- Laboratory of Nervous System Disorders and Therapies, GIGA Neurosciences, University of Liège, Liège, Belgium
| | - Brigitte Malgrange
- Laboratory of Developmental Neurobiology, GIGA Stem Cells & GIGA Neurosciences, University of Liège, Liège, Belgium
| | - Renaud Vandenbosch
- Laboratory of Developmental Neurobiology, GIGA Stem Cells & GIGA Neurosciences, University of Liège, Liège, Belgium.,Division of Histology, Department of Biomedical and Preclinical Sciences, University of Liège, Liège, Belgium
| | - Rachelle Franzen
- Laboratory of Nervous System Disorders and Therapies, GIGA Neurosciences, University of Liège, Liège, Belgium
| |
Collapse
|
14
|
David-Bercholz J, Kuo CT, Deneen B. Astrocyte and Oligodendrocyte Responses From the Subventricular Zone After Injury. Front Cell Neurosci 2022; 15:797553. [PMID: 35002630 PMCID: PMC8740317 DOI: 10.3389/fncel.2021.797553] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 11/15/2021] [Indexed: 11/14/2022] Open
Abstract
Under normal conditions, neural stem cells (NSCs or B cells) in the adult subventricular zone (SVZ) give rise to amplifying neural progenitor cells (NPCs or C cells), which can produce neuroblasts (or A cells) that migrate to the olfactory bulb and differentiate into new neurons. However, following brain injury, these cells migrate toward the injury site where they differentiate into astrocytes and oligodendrocytes. In this review, we will focus on recent findings that chronicle how astrocytes and oligodendrocytes derived from SVZ-NSCs respond to different types of injury. We will also discuss molecular regulators of SVZ-NSC proliferation and their differentiation into astrocytes and oligodendrocytes. Overall, the goal of this review is to highlight how SVZ-NSCs respond to injury and to summarize the regulatory mechanisms that oversee their glial response. These molecular and cellular processes will provide critical insights needed to develop strategies to promote brain repair following injury using SVZ-NSCs.
Collapse
Affiliation(s)
- Jennifer David-Bercholz
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, United States.,Department of Anesthesiology, Duke University School of Medicine, Durham, NC, United States
| | - Chay T Kuo
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, United States
| | - Benjamin Deneen
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, United States.,Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States.,Department of Neurosurgery, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
15
|
Wang MJ, Li ZH, Gao RW, Chen QF, Lin J, Xiao ML, Zhang K, Chen C. Effects of delayed HIF-1α expression in astrocytes on myelination following hypoxia-ischaemia white matter injury in immature rats. Transl Pediatr 2022; 11:20-32. [PMID: 35242649 PMCID: PMC8825930 DOI: 10.21037/tp-21-407] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Accepted: 12/15/2021] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND The underlying cause of neurological sequelae after immature cerebral hypoxia-ischaemia (HI) white matter injury is impaired myelination. Previous studies have indicated that astrocyte activation is closely related to impaired myelination. However, the mechanism of reactive gliosis in white matter injury post-HI remains poorly understood. METHODS Studies using adult ischaemic animal models demonstrated that hypoxia inducible factor-1α (HIF-1α) expression was involved in the formation of reactive astrocytes. Here, we investigated the temporal expression of HIF-1α and its impact on reactive gliosis and further myelination using a perinatal HI white matter injury model induced in rats at postnatal day 3. The temporal pattern of HIF-1α expression post-HI injury was tested by western blotting and immunofluorescence. Rats were treated with a HIF-1α inhibitor at 72 hours post-HI injury. Reactive gliosis and myelination were assessed with western blotting, immunofluorescence and electron microscopy, and neurological functions were examined by behavioural testing. RESULTS Our results showed that the expression of HIF-1α was upregulated in neurons at 24 hours and in astrocytes at 7 days post-HI. Inhibiting delayed HIF-1α expression post-HI injury could restrain reactive gliosis, ameliorate hypomyelination, and improve the performance of rats in the Morris water maze test. CONCLUSIONS Our findings suggest that a delayed increase in HIF-1α in astrocytes is involved in glial scar formation and leads to arrested oligodendrocyte maturation, impaired myelination, and long-term neurological function after experimental white matter injury in immature rats.
Collapse
Affiliation(s)
- Min-Jie Wang
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China.,Key Laboratory of Neonatal Disease, Ministry of Health, Shanghai, China
| | - Zhi-Hua Li
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China.,Key Laboratory of Neonatal Disease, Ministry of Health, Shanghai, China
| | - Rui-Wei Gao
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China.,Key Laboratory of Neonatal Disease, Ministry of Health, Shanghai, China
| | - Qiu-Fan Chen
- Key Laboratory of Neonatal Disease, Ministry of Health, Shanghai, China
| | - Jie Lin
- Key Laboratory of Neonatal Disease, Ministry of Health, Shanghai, China
| | - Mi-Li Xiao
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China.,Key Laboratory of Neonatal Disease, Ministry of Health, Shanghai, China
| | - Ke Zhang
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China.,Key Laboratory of Neonatal Disease, Ministry of Health, Shanghai, China
| | - Chao Chen
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China.,Key Laboratory of Neonatal Disease, Ministry of Health, Shanghai, China
| |
Collapse
|
16
|
Yazdani A, Howidi B, Shi MZ, Tugarinov N, Khoja Z, Wintermark P. Sildenafil improves hippocampal brain injuries and restores neuronal development after neonatal hypoxia-ischemia in male rat pups. Sci Rep 2021; 11:22046. [PMID: 34764335 PMCID: PMC8586032 DOI: 10.1038/s41598-021-01097-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 10/08/2021] [Indexed: 02/07/2023] Open
Abstract
The hippocampus is a fundamental structure of the brain that plays an important role in neurodevelopment and is very sensitive to hypoxia-ischemia (HI). The purpose of this study was to investigate the effects of sildenafil on neonatal hippocampal brain injuries resulting from HI, and on neuronal development in this context. HI was induced in male Long-Evans rat pups at postnatal day 10 (P10) by a left common carotid ligation followed by a 2-h exposure to 8% oxygen. Rat pups were randomized to vehicle or sildenafil given orally twice daily for 7 days starting 12 h after HI. Hematoxylin and eosin staining was performed at P30 to measure the surface of the hippocampus; immunohistochemistry was performed to stain neurons, oligodendrocytes, and glial cells in the hippocampus. Western blots of the hippocampus were performed at P12, P17, and P30 to study the expression of neuronal markers and mTOR pathway. HI caused significant hippocampal atrophy and a significant reduction of the number of mature neurons, and induced reactive astrocytosis and microgliosis in the hippocampus. HI increased apoptosis and caused significant dysregulation of the normal neuronal development program. Treatment with sildenafil preserved the gross morphology of the hippocampus, reverted the number of mature neurons to levels comparable to sham rats, significantly increased both the immature and mature oligodendrocytes, and significantly reduced the number of microglia and astrocytes. Sildenafil also decreased apoptosis and reestablished the normal progression of post-natal neuronal development. The PI3K/Akt/mTOR pathway, whose activity was decreased after HI in the hippocampus, and restored after sildenafil treatment, may be involved. Sildenafil may have both neuroprotective and neurorestorative properties in the neonatal hippocampus following HI.
Collapse
Affiliation(s)
- Armin Yazdani
- grid.63984.300000 0000 9064 4811Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Belal Howidi
- grid.63984.300000 0000 9064 4811Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Meng Zhu Shi
- grid.63984.300000 0000 9064 4811Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Nicol Tugarinov
- grid.63984.300000 0000 9064 4811Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Zehra Khoja
- grid.63984.300000 0000 9064 4811Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Pia Wintermark
- Research Institute of the McGill University Health Centre, Montreal, Canada. .,Division of Newborn Medicine, Department of Pediatrics, Montreal Children's Hospital, 1001 boul. Décarie, Site Glen Block E, EM0.3244, Montreal, QC, H4A 3J1, Canada.
| |
Collapse
|
17
|
Du S, Zheng H. Role of FoxO transcription factors in aging and age-related metabolic and neurodegenerative diseases. Cell Biosci 2021; 11:188. [PMID: 34727995 PMCID: PMC8561869 DOI: 10.1186/s13578-021-00700-7] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 10/20/2021] [Indexed: 12/18/2022] Open
Abstract
Aging happens to all of us as we live. Thanks to the improved living standard and discovery of life-saving medicines, our life expectancy has increased substantially across the world in the past century. However, the rise in lifespan leads to unprecedented increases in both the number and the percentage of individuals 65 years and older, accompanied by the increased incidences of age-related diseases such as type 2 diabetes mellitus and Alzheimer's disease. FoxO transcription factors are evolutionarily conserved molecules that play critical roles in diverse biological processes, in particular aging and metabolism. Their dysfunction is often found in the pathogenesis of many age-related diseases. Here, we summarize the signaling pathways and cellular functions of FoxO proteins. We also review the complex role of FoxO in aging and age-related diseases, with focus on type 2 diabetes and Alzheimer's disease and discuss the possibility of FoxO as a molecular link between aging and disease risks.
Collapse
Affiliation(s)
- Shuqi Du
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA
| | - Hui Zheng
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
18
|
Chen J, Guan Z. Function of Oncogene Mycn in Adult Neurogenesis and Oligodendrogenesis. Mol Neurobiol 2021; 59:77-92. [PMID: 34625907 PMCID: PMC8786763 DOI: 10.1007/s12035-021-02584-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 09/27/2021] [Indexed: 11/17/2022]
Abstract
Human MYCN is an oncogene amplified in neuroblastoma and many other tumors. Both human MYCN and mouse Mycn genes are important in embryonic brain development, but their functions in adult healthy nerve system are completely unknown. Here, with Mycn-eGFP mice and quantitative RT-PCR, we found that Mycn was expressed in specific brain regions of young adult mice, including subventricular zone (SVZ), subgranular zone (SGZ), olfactory bulb (OB), subcallosal zone (SCZ), and corpus callosum (CC). With immunohistochemistry (IHC), we found that many Mycn-expressing cells expressed neuroblast marker doublecortin (DCX) and proliferation marker Ki67. With Dcx-creER and Mki67-creER mouse lines, we fate mapped Dcx-expressing neuroblasts and Mki67-expressing proliferation cells, along with deleting Mycn from these cells in adult mice. We found that knocking out Mycn from adult neuroblasts or proliferating cells significantly reduced cells in proliferation in SVZ, SGZ, OB, SCZ, and CC. We also demonstrated that the Mycn-deficient neuroblasts in SGZ matured quicker than wild-type neuroblasts, and that Mycn-deficient proliferating cells were more likely to survive in SVZ, SGZ, OB, SCZ, and CC compared to wild type. Thus, our results demonstrate that, in addition to causing tumors in the nervous system, oncogene Mycn has a crucial function in neurogenesis and oligodendrogenesis in adult healthy brain.
Collapse
Affiliation(s)
- Jiao Chen
- Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Zhonghui Guan
- Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, CA, 94143, USA.
| |
Collapse
|
19
|
Role of macrophages in fetal development and perinatal disorders. Pediatr Res 2021; 90:513-523. [PMID: 33070164 DOI: 10.1038/s41390-020-01209-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 09/17/2020] [Accepted: 09/25/2020] [Indexed: 02/06/2023]
Abstract
In the fetus and the neonate, altered macrophage function has been implicated not only in inflammatory disorders but also in developmental abnormalities marked by altered onset, interruption, or imbalance of key structural changes. The developmental role of macrophages were first noted nearly a century ago, at about the same time when these cells were being identified as central effectors in phagocytosis and elimination of microbes. Since that time, we have made considerable progress in understanding the diverse roles that these cells play in both physiology and disease. Here, we review the role of fetal and neonatal macrophages in immune surveillance, innate immunity, homeostasis, tissue remodeling, angiogenesis, and repair of damaged tissues. We also discuss the possibility of therapeutic manipulation of the relative abundance and activation status of macrophage subsets in various diseases. This article combines peer-reviewed evidence from our own studies with results of an extensive literature search in the databases PubMed, EMBASE, and Scopus. IMPACT: We have reviewed the structure, differentiation, and classification of macrophages in the neonatal period. Neonatal macrophages are derived from embryonic, hepatic, and bone marrow precursors. Macrophages play major roles in tissue homeostasis, innate immunity, inflammation, tissue repair, angiogenesis, and apoptosis of various cellular lineages in various infectious and inflammatory disorders. Macrophages and related inflammatory mediators could be important therapeutic targets in several neonatal diseases.
Collapse
|
20
|
Allan KC, Hu LR, Scavuzzo MA, Morton AR, Gevorgyan AS, Cohn EF, Clayton BL, Bederman IR, Hung S, Bartels CF, Madhavan M, Tesar PJ. Non-canonical Targets of HIF1a Impair Oligodendrocyte Progenitor Cell Function. Cell Stem Cell 2021; 28:257-272.e11. [PMID: 33091368 PMCID: PMC7867598 DOI: 10.1016/j.stem.2020.09.019] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 08/19/2020] [Accepted: 09/29/2020] [Indexed: 12/25/2022]
Abstract
Mammalian cells respond to insufficient oxygen through transcriptional regulators called hypoxia-inducible factors (HIFs). Although transiently protective, prolonged HIF activity drives distinct pathological responses in different tissues. Using a model of chronic HIF1a accumulation in pluripotent-stem-cell-derived oligodendrocyte progenitors (OPCs), we demonstrate that HIF1a activates non-canonical targets to impair generation of oligodendrocytes from OPCs. HIF1a activated a unique set of genes in OPCs through interaction with the OPC-specific transcription factor OLIG2. Non-canonical targets, including Ascl2 and Dlx3, were sufficient to block differentiation through suppression of the oligodendrocyte regulator Sox10. Chemical screening revealed that inhibition of MEK/ERK signaling overcame the HIF1a-mediated block in oligodendrocyte generation by restoring Sox10 expression without affecting canonical HIF1a activity. MEK/ERK inhibition also drove oligodendrocyte formation in hypoxic regions of human oligocortical spheroids. This work defines mechanisms by which HIF1a impairs oligodendrocyte formation and establishes that cell-type-specific HIF1a targets perturb cell function in response to low oxygen.
Collapse
Affiliation(s)
- Kevin C. Allan
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| | - Lucille R. Hu
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| | - Marissa A. Scavuzzo
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| | - Andrew R. Morton
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| | - Artur S. Gevorgyan
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| | - Erin F. Cohn
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| | - Benjamin L.L. Clayton
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| | - Ilya R. Bederman
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| | - Stevephen Hung
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| | - Cynthia F. Bartels
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| | - Mayur Madhavan
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| | - Paul J. Tesar
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA.,Lead Contact,Correspondence:
| |
Collapse
|
21
|
HIFα Regulates Developmental Myelination Independent of Autocrine Wnt Signaling. J Neurosci 2020; 41:251-268. [PMID: 33208471 DOI: 10.1523/jneurosci.0731-20.2020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 10/15/2020] [Accepted: 11/11/2020] [Indexed: 01/17/2023] Open
Abstract
The developing CNS is exposed to physiological hypoxia, under which hypoxia-inducible factor α (HIFα) is stabilized and plays a crucial role in regulating neural development. The cellular and molecular mechanisms of HIFα in developmental myelination remain incompletely understood. A previous concept proposes that HIFα regulates CNS developmental myelination by activating the autocrine Wnt/β-catenin signaling in oligodendrocyte progenitor cells (OPCs). Here, by analyzing a battery of genetic mice of both sexes, we presented in vivo evidence supporting an alternative understanding of oligodendroglial HIFα-regulated developmental myelination. At the cellular level, we found that HIFα was required for developmental myelination by transiently controlling upstream OPC differentiation but not downstream oligodendrocyte maturation and that HIFα dysregulation in OPCs but not oligodendrocytes disturbed normal developmental myelination. We demonstrated that HIFα played a minor, if any, role in regulating canonical Wnt signaling in the oligodendroglial lineage or in the CNS. At the molecular level, blocking autocrine Wnt signaling did not affect HIFα-regulated OPC differentiation and myelination. We further identified HIFα-Sox9 regulatory axis as an underlying molecular mechanism in HIFα-regulated OPC differentiation. Our findings support a concept shift in our mechanistic understanding of HIFα-regulated CNS myelination from the previous Wnt-dependent view to a Wnt-independent one and unveil a previously unappreciated HIFα-Sox9 pathway in regulating OPC differentiation.SIGNIFICANCE STATEMENT Promoting disturbed developmental myelination is a promising option in treating diffuse white matter injury, previously called periventricular leukomalacia, a major form of brain injury affecting premature infants. In the developing CNS, hypoxia-inducible factor α (HIFα) is a key regulator that adapts neural cells to physiological and pathologic hypoxic cues. The role and mechanism of HIFα in oligodendroglial myelination, which is severely disturbed in preterm infants affected with diffuse white matter injury, is incompletely understood. Our findings presented here represent a concept shift in our mechanistic understanding of HIFα-regulated developmental myelination and suggest the potential of intervening with an oligodendroglial HIFα-mediated signaling pathway to mitigate disturbed myelination in premature white matter injury.
Collapse
|
22
|
Ma Q, Matsunaga A, Ho B, Oksenberg JR, Didonna A. Oligodendrocyte-specific Argonaute profiling identifies microRNAs associated with experimental autoimmune encephalomyelitis. J Neuroinflammation 2020; 17:297. [PMID: 33046105 PMCID: PMC7552381 DOI: 10.1186/s12974-020-01964-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 09/24/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND MicroRNAs (miRNAs) belong to a class of evolutionary conserved, non-coding small RNAs with regulatory functions on gene expression. They negatively affect the expression of target genes by promoting either RNA degradation or translational inhibition. In recent years, converging studies have identified miRNAs as key regulators of oligodendrocyte (OL) functions. OLs are the cells responsible for the formation and maintenance of myelin in the central nervous system (CNS) and represent a principal target of the autoimmune injury in multiple sclerosis (MS). METHODS MiRAP is a novel cell-specific miRNA affinity-purification technique which relies on genetically tagging Argonaut 2 (AGO2), an enzyme involved in miRNA processing. Here, we exploited miRAP potentiality to characterize OL-specific miRNA dynamics in the MS model experimental autoimmune encephalomyelitis (EAE). RESULTS We show that 20 miRNAs are differentially regulated in OLs upon transition from pre-symptomatic EAE stages to disease peak. Subsequent in vitro differentiation experiments demonstrated that a sub-group of them affects the OL maturation process, mediating either protective or detrimental signals. Lastly, transcriptome profiling highlighted the endocytosis, ferroptosis, and FoxO cascades as the pathways associated with miRNAs mediating or inhibiting OL maturation. CONCLUSIONS Altogether, our work supports a dual role for miRNAs in autoimmune demyelination. In particular, the enrichment in miRNAs mediating pro-myelinating signals suggests an active involvement of these non-coding RNAs in the homeostatic response toward neuroinflammatory injury.
Collapse
Affiliation(s)
- Qin Ma
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, 675 Nelson Rising Lane, San Francisco, CA, 94158, USA
| | - Atsuko Matsunaga
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, 675 Nelson Rising Lane, San Francisco, CA, 94158, USA
| | - Brenda Ho
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, 675 Nelson Rising Lane, San Francisco, CA, 94158, USA
| | - Jorge R Oksenberg
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, 675 Nelson Rising Lane, San Francisco, CA, 94158, USA
| | - Alessandro Didonna
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, 675 Nelson Rising Lane, San Francisco, CA, 94158, USA.
| |
Collapse
|
23
|
Hamdy N, Eide S, Sun HS, Feng ZP. Animal models for neonatal brain injury induced by hypoxic ischemic conditions in rodents. Exp Neurol 2020; 334:113457. [PMID: 32889009 DOI: 10.1016/j.expneurol.2020.113457] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 08/28/2020] [Accepted: 08/30/2020] [Indexed: 02/06/2023]
Abstract
Neonatal hypoxia-ischemia and resulting encephalopathies are of significant concern. Intrapartum asphyxia is a leading cause of neonatal death globally. Among surviving infants, there remains a high incidence of hypoxic-ischemic encephalopathy due to neonatal hypoxic-ischemic brain injury, manifesting as mild conditions including attention deficit hyperactivity disorder, and debilitating disorders such as cerebral palsy. Various animal models of neonatal hypoxic brain injury have been implemented to explore cellular and molecular mechanisms, assess the potential of novel therapeutic strategies, and characterize the functional and behavioural correlates of injury. Each of the animal models has individual advantages and limitations. The present review looks at several widely-used and alternative rodent models of neonatal hypoxia and hypoxia-ischemia; it highlights their strengths and limitations, and their potential for continued and improved use.
Collapse
Affiliation(s)
- Nancy Hamdy
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Sarah Eide
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Hong-Shuo Sun
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada; Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada.
| | - Zhong-Ping Feng
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada.
| |
Collapse
|
24
|
Srivastava T, Sherman LS, Back SA. Dysregulation of Hyaluronan Homeostasis During White Matter Injury. Neurochem Res 2020; 45:672-683. [PMID: 31542857 PMCID: PMC7060835 DOI: 10.1007/s11064-019-02879-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 09/16/2019] [Accepted: 09/16/2019] [Indexed: 12/13/2022]
Abstract
Although the extra cellular matrix (ECM) comprises a major proportion of the CNS parenchyma, new roles for the ECM in regeneration and repair responses to CNS injury have only recently been appreciated. The ECM undergoes extensive remodeling following injury to the developing or mature CNS in disorders that -include perinatal hypoxic-ischemic cerebral injury, multiple sclerosis and age-related vascular dementia. Here we focus on recently described mechanisms involving hyaluronan (HA), which negatively impact myelin repair after cerebral white matter injury. Injury induced depolymerization of hyaluronan (HA)-a component of the neural ECM-can inhibit myelin repair through the actions of specific sizes of HA fragments. These bioactive fragments selectively block the maturation of late oligodendrocyte progenitors via an immune tolerance-like pathway that suppresses pro-myelination signaling. We highlight emerging new pathophysiological roles of the neural ECM, particularly of those played by HA fragments (HAf) after injury and discuss strategies to promoter repair and regeneration of chronic myelination failure.
Collapse
Affiliation(s)
- Taasin Srivastava
- Department of Pediatrics, Oregon Health & Science University, Portland, OR, USA
| | - Larry S Sherman
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, USA
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, OR, USA
| | - Stephen A Back
- Department of Pediatrics, Oregon Health & Science University, Portland, OR, USA.
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA.
- Department of Pediatrics, Division of Pediatric Neuroscience, Oregon Health and Science University, 3181 S.W. Sam Jackson Park Rd, Portland, OR, 97239-3098, USA.
| |
Collapse
|
25
|
Forbes TA, Goldstein EZ, Dupree JL, Jablonska B, Scafidi J, Adams KL, Imamura Y, Hashimoto-Torii K, Gallo V. Environmental enrichment ameliorates perinatal brain injury and promotes functional white matter recovery. Nat Commun 2020; 11:964. [PMID: 32075970 PMCID: PMC7031237 DOI: 10.1038/s41467-020-14762-7] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 01/31/2020] [Indexed: 12/14/2022] Open
Abstract
Hypoxic damage to the developing brain due to preterm birth causes many anatomical changes, including damage to the periventricular white matter. This results in the loss of glial cells, significant disruptions in myelination, and thereby cognitive and behavioral disabilities seen throughout life. Encouragingly, these neurological morbidities can be improved by environmental factors; however, the underlying cellular mechanisms remain unknown. We found that early and continuous environmental enrichment selectively enhances endogenous repair of the developing white matter by promoting oligodendroglial maturation, myelination, and functional recovery after perinatal brain injury. These effects require increased exposure to socialization, physical activity, and cognitive enhancement of surroundings-a complete enriched environment. Using RNA-sequencing, we identified oligodendroglial-specific responses to hypoxic brain injury, and uncovered molecular mechanisms involved in enrichment-induced recovery. Together, these results indicate that myelin plasticity induced by modulation of the neonatal environment can be targeted as a therapeutic strategy for preterm birth.
Collapse
Affiliation(s)
- Thomas A Forbes
- Center for Neuroscience Research, Children's Research Institute, Children's National Hospital, Washington, DC, 20010, USA.,Institute for Biomedical Sciences, The George Washington University, Washington, DC, 20052, USA
| | - Evan Z Goldstein
- Center for Neuroscience Research, Children's Research Institute, Children's National Hospital, Washington, DC, 20010, USA
| | - Jeffrey L Dupree
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA, 23284, USA
| | - Beata Jablonska
- Center for Neuroscience Research, Children's Research Institute, Children's National Hospital, Washington, DC, 20010, USA.,Institute for Biomedical Sciences, The George Washington University, Washington, DC, 20052, USA
| | - Joseph Scafidi
- Center for Neuroscience Research, Children's Research Institute, Children's National Hospital, Washington, DC, 20010, USA.,Institute for Biomedical Sciences, The George Washington University, Washington, DC, 20052, USA
| | - Katrina L Adams
- Center for Neuroscience Research, Children's Research Institute, Children's National Hospital, Washington, DC, 20010, USA
| | - Yuka Imamura
- Institute for Personalized Medicine, Penn State University, College of Medicine, Hershey, PA, 17033, USA
| | - Kazue Hashimoto-Torii
- Center for Neuroscience Research, Children's Research Institute, Children's National Hospital, Washington, DC, 20010, USA
| | - Vittorio Gallo
- Center for Neuroscience Research, Children's Research Institute, Children's National Hospital, Washington, DC, 20010, USA. .,Institute for Biomedical Sciences, The George Washington University, Washington, DC, 20052, USA.
| |
Collapse
|
26
|
Truttmann AC, Ginet V, Puyal J. Current Evidence on Cell Death in Preterm Brain Injury in Human and Preclinical Models. Front Cell Dev Biol 2020; 8:27. [PMID: 32133356 PMCID: PMC7039819 DOI: 10.3389/fcell.2020.00027] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 01/14/2020] [Indexed: 12/19/2022] Open
Abstract
Despite tremendous advances in neonatal intensive care over the past 20 years, prematurity carries a high burden of neurological morbidity lasting lifelong. The term encephalopathy of prematurity (EoP) coined by Volpe in 2009 encompasses all aspects of the now known effects of prematurity on the immature brain, including altered and disturbed development as well as specific lesional hallmarks. Understanding the way cells are damaged is crucial to design brain protective strategies, and in this purpose, preclinical models largely contribute to improve the comprehension of the cell death mechanisms. While neuronal cell death has been deeply investigated and characterized in (hypoxic–ischemic) encephalopathy of the newborn at term, little is known about the types of cell death occurring in preterm brain injury. Three main different morphological cell death types are observed in the immature brain, specifically in models of hypoxic–ischemic encephalopathy, namely, necrotic, apoptotic, and autophagic cell death. Features of all three types may be present in the same dying neuron. In preterm brain injury, description of cell death types is sparse, and cell loss primarily concerns immature oligodendrocytes and, infrequently, neurons. In the present review, we first shortly discuss the different main severe preterm brain injury conditions that have been reported to involve cell death, including periventricular leucomalacia (PVL), diffuse white matter injury (dWMI), and intraventricular hemorrhages, as well as potentially harmful iatrogenic conditions linked to premature birth (anesthesia and caffeine therapy). Then, we present an overview of current evidence concerning cell death in both clinical human tissue data and preclinical models by focusing on studies investigating the presence of cell death allowing discriminating between the types of cell death involved. We conclude that, to improve brain protective strategies, not only apoptosis but also other cell death (such as regulated necrotic and autophagic) pathways now need to be investigated together in order to consider all cell death mechanisms involved in the pathogenesis of preterm brain damage.
Collapse
Affiliation(s)
- Anita C Truttmann
- Clinic of Neonatology, Department of Women, Mother and Child, University Hospital Center of Vaud, Lausanne, Switzerland
| | - Vanessa Ginet
- Clinic of Neonatology, Department of Women, Mother and Child, University Hospital Center of Vaud, Lausanne, Switzerland.,Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Julien Puyal
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland.,CURML, University Center of Legal Medicine, Lausanne University Hospital, Lausanne, Switzerland
| |
Collapse
|
27
|
Extrinsic Factors Driving Oligodendrocyte Lineage Cell Progression in CNS Development and Injury. Neurochem Res 2020; 45:630-642. [PMID: 31997102 PMCID: PMC7058689 DOI: 10.1007/s11064-020-02967-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 12/19/2019] [Accepted: 12/21/2019] [Indexed: 12/15/2022]
Abstract
Oligodendrocytes (OLs) generate myelin membranes for the rapid propagation of electrical signals along axons in the central nervous system (CNS) and provide metabolites to support axonal integrity and function. Differentiation of OLs from oligodendrocyte progenitor cells (OPCs) is orchestrated by a multitude of intrinsic and extrinsic factors in the CNS. Disruption of this process, or OL loss in the developing or adult brain, as observed in various neurological conditions including hypoxia/ischemia, stroke, and demyelination, results in axonal dystrophy, neuronal dysfunction, and severe neurological impairments. While much is known regarding the intrinsic regulatory signals required for OL lineage cell progression in development, studies from pathological conditions highlight the importance of the CNS environment and external signals in regulating OL genesis and maturation. Here, we review the recent findings in OL biology in the context of the CNS physiological and pathological conditions, focusing on extrinsic factors that facilitate OL development and regeneration.
Collapse
|
28
|
White Matter and Neuroprotection in Alzheimer's Dementia. Molecules 2020; 25:molecules25030503. [PMID: 31979414 PMCID: PMC7038211 DOI: 10.3390/molecules25030503] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 01/20/2020] [Accepted: 01/21/2020] [Indexed: 12/12/2022] Open
Abstract
Myelin is the main component of the white matter of the central nervous system (CNS), allowing the proper electrical function of the neurons by ensheathing and insulating the axons. The extensive use of magnetic resonance imaging has highlighted the white matter alterations in Alzheimer’s dementia (AD) and other neurodegenerative diseases, alterations which are early, extended, and regionally selective. Given that the white matter turnover is considerable in the adulthood, and that myelin repair is currently recognized as being the only true reparative capability of the mature CNS, oligodendrocyte precursor cells (OPCs), the cells that differentiate in oligodendrocyte, responsible for myelin formation and repair, are regarded as a potential target for neuroprotection. In this review, several aspects of the OPC biology are reviewed. The histology and functional role of OPCs in the neurovascular-neuroglial unit as described in preclinical and clinical studies on AD is discussed, such as the OPC vulnerability to hypoxia-ischemia, neuroinflammation, and amyloid deposition. Finally, the position of OPCs in drug discovery strategies for dementia is discussed.
Collapse
|
29
|
Glial Factors Regulating White Matter Development and Pathologies of the Cerebellum. Neurochem Res 2020; 45:643-655. [PMID: 31974933 PMCID: PMC7058568 DOI: 10.1007/s11064-020-02961-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 01/07/2020] [Accepted: 01/09/2020] [Indexed: 12/31/2022]
Abstract
The cerebellum is a brain region that undergoes extremely dynamic growth during perinatal and postnatal development which is regulated by the proper interaction between glial cells and neurons with a complex concert of growth factors, chemokines, cytokines, neurotransmitters and transcriptions factors. The relevance of cerebellar functions for not only motor performance but also for cognition, emotion, memory and attention is increasingly being recognized and acknowledged. Since perturbed circuitry of cerebro-cerebellar trajectories can play a role in many central nervous system pathologies and thereby contribute to neurological symptoms in distinct neurodevelopmental and neurodegenerative diseases, is it the aim with this mini-review to highlight the pathways of glia–glia interplay being involved. The designs of future treatment strategies may hence be targeted to molecular pathways also playing a role in development and disease of the cerebellum.
Collapse
|
30
|
Baldassarro VA, Marchesini A, Giardino L, Calzà L. Differential effects of glucose deprivation on the survival of fetal versus adult neural stem cells-derived oligodendrocyte precursor cells. Glia 2019; 68:898-917. [PMID: 31755592 DOI: 10.1002/glia.23750] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 10/28/2019] [Accepted: 10/29/2019] [Indexed: 12/27/2022]
Abstract
Impaired myelination is a key feature in neonatal hypoxia/ischemia (HI), the most common perinatal/neonatal cause of death and permanent disabilities, which is triggered by the establishment of an inflammatory and hypoxic environment during the most critical period of myelin development. This process is dependent on oligodendrocyte precursor cells (OPCs) and their capability to differentiate into mature oligodendrocytes. In this study, we investigated the vulnerability of fetal and adult OPCs derived from neural stem cells (NSCs) to inflammatory and HI insults. The resulting OPCs/astrocytes cultures were exposed to cytokines to mimic inflammation, or to oxygen-glucose deprivation (OGD) to mimic an HI condition. The differentiation of both fetal and adult OPCs is completely abolished following exposure to inflammatory cytokines, while only fetal-derived OPCs degenerate when exposed to OGD. We then investigated possible mechanisms involved in OGD-mediated toxicity: (a) T3-mediated maturation induction; (b) glutamate excitotoxicity; (c) glucose metabolism. We found that while no substantial differences were observed in T3 intracellular content regulation and glutamate-mediated toxicity, glucose deprivation lead to selective OPC cell death and impaired differentiation in fetal cultures only. These results indicate that the biological response of OPCs to inflammation and demyelination is different in fetal and adult cells, and that the glucose metabolism perturbation in fetal central nervous system (CNS) may significantly contribute to neonatal pathologies. An understanding of the underlying molecular mechanism will contribute greatly to differentiating myelination enhancing and neuroprotective therapies for neonatal and adult CNS white matter lesions.
Collapse
Affiliation(s)
- Vito Antonio Baldassarro
- Health Science and Technologies Interdepartmental Center for Industrial Research (HST-ICIR), University of Bologna, Bologna, Italy.,Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy.,Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France
| | | | - Luciana Giardino
- Health Science and Technologies Interdepartmental Center for Industrial Research (HST-ICIR), University of Bologna, Bologna, Italy.,IRET Foundation, Ozzano Emilia, Italy.,Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | - Laura Calzà
- Health Science and Technologies Interdepartmental Center for Industrial Research (HST-ICIR), University of Bologna, Bologna, Italy.,Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy.,IRET Foundation, Ozzano Emilia, Italy
| |
Collapse
|
31
|
The functional microscopic neuroanatomy of the human subthalamic nucleus. Brain Struct Funct 2019; 224:3213-3227. [PMID: 31562531 PMCID: PMC6875153 DOI: 10.1007/s00429-019-01960-3] [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: 06/06/2019] [Accepted: 09/14/2019] [Indexed: 01/19/2023]
Abstract
The subthalamic nucleus (STN) is successfully used as a surgical target for deep brain stimulation in the treatment of movement disorders. Interestingly, the internal structure of the STN is still incompletely understood. The objective of the present study was to investigate three-dimensional (3D) immunoreactivity patterns for 12 individual protein markers for GABA-ergic, serotonergic, dopaminergic as well as glutamatergic signaling. We analyzed the immunoreactivity using optical densities and created a 3D reconstruction of seven postmortem human STNs. Quantitative modeling of the reconstructed 3D immunoreactivity patterns revealed that the applied protein markers show a gradient distribution in the STN. These gradients were predominantly organized along the ventromedial to dorsolateral axis of the STN. The results are of particular interest in view of the theoretical underpinning for surgical targeting, which is based on a tripartite distribution of cognitive, limbic and motor function in the STN.
Collapse
|
32
|
Romanowicz J, Leonetti C, Dhari Z, Korotcova L, Ramachandra SD, Saric N, Morton PD, Bansal S, Cheema A, Gallo V, Jonas RA, Ishibashi N. Treatment With Tetrahydrobiopterin Improves White Matter Maturation in a Mouse Model for Prenatal Hypoxia in Congenital Heart Disease. J Am Heart Assoc 2019; 8:e012711. [PMID: 31331224 PMCID: PMC6761654 DOI: 10.1161/jaha.119.012711] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 06/24/2019] [Indexed: 01/05/2023]
Abstract
Background Reduced oxygen delivery in congenital heart disease causes delayed brain maturation and white matter abnormalities in utero. No treatment currently exists. Tetrahydrobiopterin (BH4) is a cofactor for neuronal nitric oxide synthase. BH4 availability is reduced upon NOS activation, such as during hypoxic conditions, and leads to toxin production. We hypothesize that BH4 levels are depleted in the hypoxic brain and that BH4 replacement therapy mitigates the toxic effects of hypoxia on white matter. Methods and Results Transgenic mice were used to visualize oligodendrocytes. Hypoxia was introduced during a period of white matter development equivalent to the human third trimester. BH4 was administered during hypoxia. BH4 levels were depleted in the hypoxic brain by direct quantification (n=7-12). The proliferation (n=3-6), apoptosis (n=3-6), and developmental stage (n=5-8) of oligodendrocytes were determined immunohistologically. Total oligodendrocytes increased after hypoxia, consistent with hypoxia-induced proliferation seen previously; however, mature oligodendrocytes were less prevalent in hypoxia, and there was accumulation of immature oligodendrocytes. BH4 treatment improved the mature oligodendrocyte number such that it did not differ from normoxia, and accumulation of immature oligodendrocytes was not observed. These results persisted beyond the initial period of hypoxia (n=3-4). Apoptosis increased with hypoxia but decreased with BH4 treatment to normoxic levels. White matter myelin levels decreased following hypoxia by western blot. BH4 treatment normalized myelination (n=6-10). Hypoxia worsened sensory-motor coordination on balance beam tasks, and BH4 therapy normalized performance (n=5-9). Conclusions Suboptimal BH4 levels influence hypoxic white matter abnormalities. Repurposing BH4 for use during fetal brain development may limit white matter dysmaturation in congenital heart disease.
Collapse
Affiliation(s)
- Jennifer Romanowicz
- Children's National Heart InstituteChildren's National Health SystemWashingtonDC
| | - Camille Leonetti
- Children's National Heart InstituteChildren's National Health SystemWashingtonDC
- Center for Neuroscience ResearchChildren's National Health SystemWashingtonDC
| | - Zaenab Dhari
- Children's National Heart InstituteChildren's National Health SystemWashingtonDC
- Center for Neuroscience ResearchChildren's National Health SystemWashingtonDC
| | - Ludmila Korotcova
- Children's National Heart InstituteChildren's National Health SystemWashingtonDC
- Center for Neuroscience ResearchChildren's National Health SystemWashingtonDC
| | - Shruti D. Ramachandra
- Children's National Heart InstituteChildren's National Health SystemWashingtonDC
- Center for Neuroscience ResearchChildren's National Health SystemWashingtonDC
| | - Nemanja Saric
- Children's National Heart InstituteChildren's National Health SystemWashingtonDC
- Center for Neuroscience ResearchChildren's National Health SystemWashingtonDC
| | - Paul D. Morton
- Children's National Heart InstituteChildren's National Health SystemWashingtonDC
- Center for Neuroscience ResearchChildren's National Health SystemWashingtonDC
| | - Shivani Bansal
- Lombardi Comprehensive Cancer CenterGeorgetown University Medical CenterWashingtonDC
| | - Amrita Cheema
- Lombardi Comprehensive Cancer CenterGeorgetown University Medical CenterWashingtonDC
| | - Vittorio Gallo
- Center for Neuroscience ResearchChildren's National Health SystemWashingtonDC
| | - Richard A. Jonas
- Children's National Heart InstituteChildren's National Health SystemWashingtonDC
- Center for Neuroscience ResearchChildren's National Health SystemWashingtonDC
| | - Nobuyuki Ishibashi
- Children's National Heart InstituteChildren's National Health SystemWashingtonDC
- Center for Neuroscience ResearchChildren's National Health SystemWashingtonDC
| |
Collapse
|
33
|
Arteaga Cabeza O, Mikrogeorgiou A, Kannan S, Ferriero DM. Advanced nanotherapies to promote neuroregeneration in the injured newborn brain. Adv Drug Deliv Rev 2019; 148:19-37. [PMID: 31678359 DOI: 10.1016/j.addr.2019.10.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 09/19/2019] [Accepted: 10/23/2019] [Indexed: 12/16/2022]
Abstract
Neonatal brain injury affects thousands of babies each year and may lead to long-term and permanent physical and neurological problems. Currently, therapeutic hypothermia is standard clinical care for term newborns with moderate to severe neonatal encephalopathy. Nevertheless, it is not completely protective, and additional strategies to restore and promote regeneration are urgently needed. One way to ensure recovery following injury to the immature brain is to augment endogenous regenerative pathways. However, novel strategies such as stem cell therapy, gene therapies and nanotechnology have not been adequately explored in this unique age group. In this perspective review, we describe current efforts that promote neuroprotection and potential targets that are unique to the developing brain, which can be leveraged to facilitate neuroregeneration.
Collapse
|
34
|
Sergi C. EPAS 1, congenital heart disease, and high altitude: disclosures by genetics, bioinformatics, and experimental embryology. Biosci Rep 2019; 39:BSR20182197. [PMID: 31015364 PMCID: PMC6509053 DOI: 10.1042/bsr20182197] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 04/04/2019] [Accepted: 04/17/2019] [Indexed: 02/07/2023] Open
Abstract
The high-altitude environment is a challenge for human settlement. Low oxygen concentrations, extreme cold, and a harsh arid climate are doubtlessly challenges for the colonization of the Tibetan plateau. I am delighted to comment on the article of Pan et al. (2018) on mutations in endothelial PAS domain-containing protein 1 (EPAS1) in congenital heart disease in Tibetans. In humans, the EPAS1 gene is responsible for coding EPAS1 protein, an alias of which is HIF2α, an acronym for hypoxia-inducible factor 2 alpha. EPAS1 is a type of hypoxia-inducible factors, which are collected as a group of transcription factors involved in body response to oxygen level. EPAS1 gene is active under hypoxic conditions and plays an essential role in the development of the heart and in the management of the catecholamine balance, mutations of which have been identified in neuroendocrine tumors. In this article, Pan et al. investigated Tibetan patients with and without non-syndromic congenital heart disease. They identified two novel EPAS1 gene mutations, of which N203H mutation significantly affected the transcription activity of the vascular endothelial growth factor (VEGF) promoter, particularly in situations of hypoxia. VEGF is a downstream target of HIF-2 (other than HIF-1), and the expression levels of either HIF-1α or HIF-2α correlate positively to VEGF expression. Pan et al.'s data may be of incitement to further evaluate protein-protein interaction and using experimental animal models. Moreover, it may also be a stimulus for setting up genetic epidemiologic studies for other populations living at high altitudes.
Collapse
Affiliation(s)
- Consolato Sergi
- National '111' Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, P.R. China
- Department of Orthopedics, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, Hubei, P.R. China
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada
- Stollery Children's Hospital, University Alberta Hospital, Edmonton, AB, Canada
| |
Collapse
|
35
|
Singh DK, Ling EA, Kaur C. Hypoxia and myelination deficits in the developing brain. Int J Dev Neurosci 2018; 70:3-11. [PMID: 29964158 DOI: 10.1016/j.ijdevneu.2018.06.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 05/28/2018] [Accepted: 06/26/2018] [Indexed: 12/15/2022] Open
Abstract
Myelination is a complex and orderly process during brain development that is essential for normal motor, cognitive and sensory functions. Cellular and molecular interactions between myelin-forming oligodendrocytes and axons are required for normal myelination in the developing brain. Oligodendrocyte progenitor cells (OPCs) proliferate and differentiate into mature myelin-forming oligodendrocytes. In this connection, astrocytes and microglia are also involved in survival and proliferation of OPCs. Hypoxic insults during the perinatal period affect the normal development, differentiation and maturation of the OPCs or cause their death resulting in impaired myelination. Several factors such as augmented release of proinflammatory cytokines by activated microglia and astrocytes, extracellular accumulation of excess glutamate and increased levels of nitric oxide are some of the underlying factors for hypoxia induced damage to the OPCs. Additionally, hypoxia also leads to down-regulation of several genes involved in oligodendrocyte differentiation encoding proteolipid protein, platelet-derived growth factor receptor and myelin-associated glycoprotein in the developing brain. Furthermore, oligodendrocytes may also accumulate increased amounts of iron in hypoxic conditions that triggers endoplasmic reticulum stress, misfolding of proteins and generation of reactive oxygen species that ultimately would lead to myelination deficits. More in-depth studies to elucidate the pathophysiological mechanisms underlying the inability of oligodendrocytes to myelinate the developing brain in hypoxic insults are desirable to develop new therapeutic options or strategies for myelination deficits.
Collapse
Affiliation(s)
- Dhiraj Kumar Singh
- Department of Anatomy, Yong Loo Lin School of Medicine, MD10, 4 Medical drive, National University of Singapore, 117597, Singapore
| | - Eng-Ang Ling
- Department of Anatomy, Yong Loo Lin School of Medicine, MD10, 4 Medical drive, National University of Singapore, 117597, Singapore
| | - Charanjit Kaur
- Department of Anatomy, Yong Loo Lin School of Medicine, MD10, 4 Medical drive, National University of Singapore, 117597, Singapore.
| |
Collapse
|
36
|
Dillenburg A, Ireland G, Holloway RK, Davies CL, Evans FL, Swire M, Bechler ME, Soong D, Yuen TJ, Su GH, Becher JC, Smith C, Williams A, Miron VE. Activin receptors regulate the oligodendrocyte lineage in health and disease. Acta Neuropathol 2018; 135:887-906. [PMID: 29397421 PMCID: PMC5954071 DOI: 10.1007/s00401-018-1813-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 01/16/2018] [Accepted: 01/29/2018] [Indexed: 12/12/2022]
Abstract
The most prevalent neurological disorders of myelin include perinatal brain injury leading to cerebral palsy in infants and multiple sclerosis in adults. Although these disorders have distinct etiologies, they share a common neuropathological feature of failed progenitor differentiation into myelin-producing oligodendrocytes and lack of myelin, for which there is an unmet clinical need. Here, we reveal that a molecular pathology common to both disorders is dysregulation of activin receptors and that activin receptor signaling is required for the majority of myelin generation in development and following injury. Using a constitutive conditional knockout of all activin receptor signaling in oligodendrocyte lineage cells, we discovered this signaling to be required for myelination via regulation of oligodendrocyte differentiation and myelin compaction. These processes were found to be dependent on the activin receptor subtype Acvr2a, which is expressed during oligodendrocyte differentiation and axonal ensheathment in development and following myelin injury. During efficient myelin regeneration, Acvr2a upregulation was seen to coincide with downregulation of Acvr2b, a receptor subtype with relatively higher ligand affinity; Acvr2b was shown to be dispensable for activin receptor-driven oligodendrocyte differentiation and its overexpression was sufficient to impair the abovementioned ligand-driven responses. In actively myelinating or remyelinating areas of human perinatal brain injury and multiple sclerosis tissue, respectively, oligodendrocyte lineage cells expressing Acvr2a outnumbered those expressing Acvr2b, whereas in non-repairing lesions Acvr2b+ cells were increased. Thus, we propose that following human white matter injury, this increase in Acvr2b expression would sequester ligand and consequently impair Acvr2a-driven oligodendrocyte differentiation and myelin formation. Our results demonstrate dysregulated activin receptor signaling in common myelin disorders and reveal Acvr2a as a novel therapeutic target for myelin generation following injury across the lifespan.
Collapse
|
37
|
Repeated exposure to sucrose for procedural pain in mouse pups leads to long-term widespread brain alterations. Pain 2018; 158:1586-1598. [PMID: 28715355 DOI: 10.1097/j.pain.0000000000000961] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Oral sucrose is administered routinely to reduce pain of minor procedures in premature infants and is recommended as standard care in international guidelines. No human or animal studies on effects of early repeated sucrose exposure on long-term brain development have been done in the context of pain. We evaluated the effects of repeated neonatal sucrose treatment before an intervention on long-term brain structure in mouse pups. Neonatal C57Bl/6J mice (n = 109) were randomly assigned to one of 2 treatments (vehicle vs sucrose) and one of 3 interventions (handling, touch, or needle-prick). Mice received 10 interventions daily from postnatal day 1 to 6 (P1-6). A dose of vehicle or 24% sucrose was given orally 2 minutes before each intervention. At P85-95, brains were scanned using a multichannel 7.0 T MRI. Volumes of 159 independent brain regions were obtained. Early repetitive sucrose exposure in mice (after correcting for whole brain volume and multiple comparisons) lead to smaller white matter volumes in the corpus callosum, stria terminalis, and fimbria (P < 0.0001). Cortical and subcortical gray matter was also affected by sucrose with smaller volumes of hippocampus and cerebellum (P < 0.0001). These significant changes in adult brain were found irrespective of the type of intervention in the neonatal period. This study provides the first evidence of long-term adverse effects of repetitive sucrose exposure and raises concerns for the use of this standard pain management practice during a period of rapid brain development in very preterm infants.
Collapse
|
38
|
Srivastava T, Diba P, Dean JM, Banine F, Shaver D, Hagen M, Gong X, Su W, Emery B, Marks DL, Harris EN, Baggenstoss B, Weigel PH, Sherman LS, Back SA. A TLR/AKT/FoxO3 immune tolerance-like pathway disrupts the repair capacity of oligodendrocyte progenitors. J Clin Invest 2018; 128:2025-2041. [PMID: 29664021 DOI: 10.1172/jci94158] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 02/28/2018] [Indexed: 12/12/2022] Open
Abstract
Cerebral white matter injury (WMI) persistently disrupts myelin regeneration by oligodendrocyte progenitor cells (OPCs). We identified a specific bioactive hyaluronan fragment (bHAf) that downregulates myelin gene expression and chronically blocks OPC maturation and myelination via a tolerance-like mechanism that dysregulates pro-myelination signaling via AKT. Desensitization of AKT occurs via TLR4 but not TLR2 or CD44. OPC differentiation was selectively blocked by bHAf in a maturation-dependent fashion at the late OPC (preOL) stage by a noncanonical TLR4/TRIF pathway that induced persistent activation of the FoxO3 transcription factor downstream of AKT. Activated FoxO3 selectively localized to oligodendrocyte lineage cells in white matter lesions from human preterm neonates and adults with multiple sclerosis. FoxO3 constraint of OPC maturation was bHAf dependent, and involved interactions at the FoxO3 and MBP promoters with the chromatin remodeling factor Brg1 and the transcription factor Olig2, which regulate OPC differentiation. WMI has adapted an immune tolerance-like mechanism whereby persistent engagement of TLR4 by bHAf promotes an OPC niche at the expense of myelination by engaging a FoxO3 signaling pathway that chronically constrains OPC differentiation.
Collapse
Affiliation(s)
- Taasin Srivastava
- Department of Pediatrics, Oregon Health & Science University (OHSU), Portland, Oregon, USA
| | - Parham Diba
- Department of Pediatrics, Oregon Health & Science University (OHSU), Portland, Oregon, USA
| | - Justin M Dean
- Department of Pediatrics, Oregon Health & Science University (OHSU), Portland, Oregon, USA
| | - Fatima Banine
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, Oregon, USA
| | - Daniel Shaver
- Department of Pediatrics, Oregon Health & Science University (OHSU), Portland, Oregon, USA
| | - Matthew Hagen
- Department of Pediatrics, Oregon Health & Science University (OHSU), Portland, Oregon, USA
| | - Xi Gong
- Department of Pediatrics, Oregon Health & Science University (OHSU), Portland, Oregon, USA
| | - Weiping Su
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, Oregon, USA
| | - Ben Emery
- Department of Neurology, OHSU, Portland, Oregon, USA
| | - Daniel L Marks
- Department of Pediatrics, Oregon Health & Science University (OHSU), Portland, Oregon, USA
| | - Edward N Harris
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| | - Bruce Baggenstoss
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Paul H Weigel
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Larry S Sherman
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, Oregon, USA.,Department of Cell, Developmental and Cancer Biology, OHSU, Portland, Oregon, USA
| | - Stephen A Back
- Department of Pediatrics, Oregon Health & Science University (OHSU), Portland, Oregon, USA.,Department of Neurology, OHSU, Portland, Oregon, USA
| |
Collapse
|
39
|
Nguyen V, Sabeur K, Maltepe E, Ameri K, Bayraktar O, Rowitch DH. Sonic Hedgehog Agonist Protects Against Complex Neonatal Cerebellar Injury. CEREBELLUM (LONDON, ENGLAND) 2018; 17:213-227. [PMID: 29134361 PMCID: PMC5849674 DOI: 10.1007/s12311-017-0895-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The cerebellum undergoes rapid growth during the third trimester and is vulnerable to injury and deficient growth in infants born prematurely. Factors associated with preterm cerebellar hypoplasia include chronic lung disease and postnatal glucocorticoid administration. We modeled chronic hypoxemia and glucocorticoid administration in neonatal mice to study whole cerebellar and cell type-specific effects of dual exposure. Chronic neonatal hypoxia resulted in permanent cerebellar hypoplasia. This was compounded by administration of prednisolone as shown by greater volume loss and Purkinje cell death. In the setting of hypoxia and prednisolone, administration of a small molecule Smoothened-Hedgehog agonist (SAG) preserved cerebellar volume and protected against Purkinje cell death. Such protective effects were observed even when SAG was given as a one-time dose after dual insult. To model complex injury and determine cell type-specific roles for the hypoxia inducible factor (HIF) pathway, we performed conditional knockout of von Hippel Lindau (VHL) to hyperactivate HIF1α in cerebellar granule neuron precursors (CGNP) or Purkinje cells. Surprisingly, HIF activation in either cell type resulted in no cerebellar deficit. However, in mice administered prednisolone, HIF overactivation in CGNPs resulted in significant cerebellar hypoplasia, whereas HIF overactivation in Purkinje cells caused cell death. Together, these findings indicate that HIF primes both cell types for injury via glucocorticoids, and that hypoxia/HIF + postnatal glucocorticoid administration act on distinct cellular pathways to cause cerebellar injury. They further suggest that SAG is neuroprotective in the setting of complex neonatal cerebellar injury.
Collapse
Affiliation(s)
- Vien Nguyen
- Department of Pediatrics, Eli and Edythe Broad Institute for Stem Cell Research and Regenerative Medicine, University of California, San Francisco, 513 Parnassus Avenue, San Francisco, CA, 94143, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, 513 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Khalida Sabeur
- Department of Pediatrics, Eli and Edythe Broad Institute for Stem Cell Research and Regenerative Medicine, University of California, San Francisco, 513 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Emin Maltepe
- Division of Neonatology, University of California, San Francisco, 513 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Kurosh Ameri
- Department of Cardiology, University of California, San Francisco, 513 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Omer Bayraktar
- Department of Pediatrics, Eli and Edythe Broad Institute for Stem Cell Research and Regenerative Medicine, University of California, San Francisco, 513 Parnassus Avenue, San Francisco, CA, 94143, USA
- Department of Paediatrics, Wellcome Trust-MRC Stem Cell Institute, Cambridge University, Cambridge, UK
| | - David H Rowitch
- Department of Pediatrics, Eli and Edythe Broad Institute for Stem Cell Research and Regenerative Medicine, University of California, San Francisco, 513 Parnassus Avenue, San Francisco, CA, 94143, USA.
- Biomedical Sciences Graduate Program, University of California, San Francisco, 513 Parnassus Avenue, San Francisco, CA, 94143, USA.
- Division of Neonatology, University of California, San Francisco, 513 Parnassus Avenue, San Francisco, CA, 94143, USA.
- Department of Paediatrics, Wellcome Trust-MRC Stem Cell Institute, Cambridge University, Cambridge, UK.
| |
Collapse
|
40
|
Kirschen GW, Kéry R, Liu H, Ahamad A, Chen L, Akmentin W, Kumar R, Levine J, Xiong Q, Ge S. Genetic dissection of the neuro-glio-vascular machinery in the adult brain. Mol Brain 2018; 11:2. [PMID: 29335006 PMCID: PMC5769320 DOI: 10.1186/s13041-017-0345-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 12/21/2017] [Indexed: 12/30/2022] Open
Abstract
The adult brain actively controls its metabolic homeostasis via the circulatory system at the blood brain barrier interface. The mechanisms underlying the functional coupling from neuron to vessel remain poorly understood. Here, we established a novel method to genetically isolate the individual components of this coupling machinery using a combination of viral vectors. We first discovered a surprising non-uniformity of the glio-vascular structure in different brain regions. We carried out a viral injection screen and found that intravenous Canine Adenovirus 2 (CAV2) preferentially targeted perivascular astrocytes throughout the adult brain, with sparing of the hippocampal hilus from infection. Using this new intravenous method to target astrocytes, we selectively ablated these cells and observed severe defects in hippocampus-dependent contextual memory and the metabolically regulated process of hippocampal neurogenesis. Combined with AAV9 targeting of neurons and endothelial cells, all components of the neuro-glio-vascular machinery can be simultaneously labeled for genetic manipulation. Together, we demonstrate a novel method, which we term CATNAP (CAV/AAV Targeting of Neurons and Astrocytes Perivascularly), to target and manipulate the neuro-glio-vascular machinery in the adult brain.
Collapse
Affiliation(s)
- Gregory W Kirschen
- Medical Scientist Training Program, Stony Brook, New York, USA.,Department of Neurobiology & Behavior, SUNY Stony Brook, Stony Brook, NY, 11794, USA
| | - Rachel Kéry
- Medical Scientist Training Program, Stony Brook, New York, USA.,Department of Neurobiology & Behavior, SUNY Stony Brook, Stony Brook, NY, 11794, USA
| | - Hanxiao Liu
- Department of Neurobiology & Behavior, SUNY Stony Brook, Stony Brook, NY, 11794, USA
| | - Afrinash Ahamad
- School of Health Technology & Management, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Liang Chen
- Department of Neurobiology & Behavior, SUNY Stony Brook, Stony Brook, NY, 11794, USA
| | - Wendy Akmentin
- Department of Neurobiology & Behavior, SUNY Stony Brook, Stony Brook, NY, 11794, USA
| | - Ramya Kumar
- Department of Neurobiology & Behavior, SUNY Stony Brook, Stony Brook, NY, 11794, USA
| | - Joel Levine
- Department of Neurobiology & Behavior, SUNY Stony Brook, Stony Brook, NY, 11794, USA
| | - Qiaojie Xiong
- Department of Neurobiology & Behavior, SUNY Stony Brook, Stony Brook, NY, 11794, USA.
| | - Shaoyu Ge
- Department of Neurobiology & Behavior, SUNY Stony Brook, Stony Brook, NY, 11794, USA.
| |
Collapse
|
41
|
Pediatric brain repair from endogenous neural stem cells of the subventricular zone. Pediatr Res 2018; 83:385-396. [PMID: 29028220 DOI: 10.1038/pr.2017.261] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 10/05/2017] [Indexed: 12/22/2022]
Abstract
There is great interest in the regenerative potential of the neural stem cells and progenitors that populate the germinal zones of the immature brain. Studies using animal models of pediatric brain injuries have provided a clearer understanding of the responses of these progenitors to injury. In this review, we have compared and contrasted the responses of the endogenous neural stem cells and progenitors of the subventricular zone in animal models of neonatal cerebral hypoxia-ischemia, neonatal stroke, congenital cardiac disease, and pediatric traumatic brain injury. We have reviewed the dynamic shifts that occur within this germinal zone with injury as well as changes in known signaling molecules that affect these progenitors. Importantly, we have summarized data on the extent to which cell replacement occurs in response to each of these injuries, opportunities available, and obstacles that will need to be overcome to improve neurological outcomes in survivors.
Collapse
|
42
|
Systemic inflammation combined with neonatal cerebellar haemorrhage aggravates long-term structural and functional outcomes in a mouse model. Brain Behav Immun 2017; 66:257-276. [PMID: 28755859 DOI: 10.1016/j.bbi.2017.07.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 07/11/2017] [Accepted: 07/19/2017] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Despite the increased recognition of cerebellar injury in survivors of preterm birth, the neurodevelopmental consequences of isolated cerebellar injury have been largely unexplored and our current understanding of the functional deficits requires further attention in order to translate knowledge to best practices. Preterm infants are exposed to multiple stressors during their postnatal development including perinatal cerebellar haemorrhage (CBH) and postnatal infection, two major risk factors for neurodevelopmental impairments. METHODS We developed a translational mouse model of CBH and/or inflammation to measure the short- and long-term outcomes in cerebellar structure and function. RESULTS Mice exposed to early combined insults of CBH and early inflammatory state (EIS) have a delay in grasping acquisition, neonatal motor deficits and deficient long-term memory. CBH combined with late inflammatory state (LIS) does not induce neonatal motor problems but leads to poor fine motor function and long-term memory deficits at adulthood. Early combined insults result in poor cerebellar growth from postnatal day 15 until adulthood shown by MRI, which are reflected in diminished volumes of cerebellar structures. There are also decreases in volumes of gray matter and hippocampus. Cerebellar microgliosis appears 24h after the combined insults and persists until postnatal day 15 in the cerebellar molecular layer and cerebellar nuclei in association with a disrupted patterning of myelin deposition, a delay of oligodendrocyte maturation and reduced white matter cerebellar volume. CONCLUSIONS Together, these findings reveal poor outcomes in developing brains exposed to combined cerebellar perinatal insults in association with cerebellar hypoplasia, persistence of microgliosis and alterations of cerebellar white matter maturation and growth.
Collapse
|
43
|
Newville J, Jantzie LL, Cunningham LA. Embracing oligodendrocyte diversity in the context of perinatal injury. Neural Regen Res 2017; 12:1575-1585. [PMID: 29171412 PMCID: PMC5696828 DOI: 10.4103/1673-5374.217320] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/12/2017] [Indexed: 12/18/2022] Open
Abstract
Emerging evidence is fueling a new appreciation of oligodendrocyte diversity that is overturning the traditional view that oligodendrocytes are a homogenous cell population. Oligodendrocytes of distinct origins, maturational stages, and regional locations may differ in their functional capacity or susceptibility to injury. One of the most unique qualities of the oligodendrocyte is its ability to produce myelin. Myelin abnormalities have been ascribed to a remarkable array of perinatal brain injuries, with concomitant oligodendrocyte dysregulation. Within this review, we discuss new insights into the diversity of the oligodendrocyte lineage and highlight their relevance in paradigms of perinatal brain injury. Future therapeutic development will be informed by comprehensive knowledge of oligodendrocyte pathophysiology that considers the particular facets of heterogeneity that this lineage exhibits.
Collapse
Affiliation(s)
- Jessie Newville
- Department of Neurosciences, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Lauren L. Jantzie
- Department of Neurosciences, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
- Department of Pediatrics, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Lee Anna Cunningham
- Department of Neurosciences, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| |
Collapse
|
44
|
Replication of JC Virus DNA in the G144 Oligodendrocyte Cell Line Is Dependent Upon Akt. J Virol 2017; 91:JVI.00735-17. [PMID: 28768870 DOI: 10.1128/jvi.00735-17] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 07/26/2017] [Indexed: 12/24/2022] Open
Abstract
Progressive multifocal leukoencephalopathy (PML) is an often-fatal demyelinating disease of the central nervous system. PML results when oligodendrocytes within immunocompromised individuals are infected with the human JC virus (JCV). We have identified an oligodendrocyte precursor cell line, termed G144, that supports robust levels of JCV DNA replication, a central part of the JCV life cycle. In addition, we have determined that JC virus readily infects G144 cells. Furthermore, we have determined that JCV DNA replication in G144 cells is stimulated by myristoylated (i.e., constitutively active) Akt and reduced by the Akt-specific inhibitor MK2206. Thus, this oligodendrocyte-based model system will be useful for a number of purposes, such as studies of JCV infection, establishing key pathways needed for the regulation of JCV DNA replication, and identifying inhibitors of this process.IMPORTANCE The disease progressive multifocal leukoencephalopathy (PML) is caused by the infection of particular brain cells, termed oligodendrocytes, by the JC virus. Studies of PML, however, have been hampered by the lack of an immortalized human cell line derived from oligodendrocytes. Here, we report that the G144 oligodendrocyte cell line supports both infection by JC virus and robust levels of JCV DNA replication. Moreover, we have established that the Akt pathway regulates JCV DNA replication and that JCV DNA replication can be inhibited by MK2206, a compound that is specific for Akt. These and related findings suggest that we have established a powerful oligodendrocyte-based model system for studies of JCV-dependent PML.
Collapse
|
45
|
Clayton BL, Huang A, Kunjamma RB, Solanki A, Popko B. The Integrated Stress Response in Hypoxia-Induced Diffuse White Matter Injury. J Neurosci 2017; 37:7465-7480. [PMID: 28720571 PMCID: PMC5546113 DOI: 10.1523/jneurosci.2738-16.2017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 06/02/2017] [Accepted: 06/25/2017] [Indexed: 12/13/2022] Open
Abstract
Currently no treatments exist for preterm infants with diffuse white matter injury (DWMI) caused by hypoxia. Due to the improved care of preterm neonates and increased recognition by advanced imaging techniques, the prevalence of DWMI is increasing. A better understanding of the pathophysiology of DWMI is therefore of critical importance. The integrated stress response (ISR), a conserved eukaryotic response to myriad stressors including hypoxia, may play a role in hypoxia-induced DWMI and may represent a novel target for much needed therapies. In this study, we use in vitro and in vivo hypoxic models of DWMI to investigate whether the ISR is involved in DWMI. We demonstrate that hypoxia activates the ISR in primary mouse oligodendrocyte precursor cells (OPCs) in vitro and that genetically inhibiting the ISR in differentiating OPCs increases their susceptibility to in vitro hypoxia. We also show that a well established in vivo mild chronic hypoxia (MCH) mouse model and a new severe acute hypoxia (SAH) mouse model of DWMI activates the initial step of the ISR. Nonetheless, genetic inhibition of the ISR has no detectable effect on either MCH- or SAH-induced DWMI. In addition, we demonstrate that genetic enhancement of the ISR does not ameliorate MCH- or SAH-induced DWMI. These studies suggest that, while the ISR protects OPCs from hypoxia in vitro, it does not appear to play a major role in either MCH- or SAH-induced DWMI and is therefore not a likely target for therapies aimed at improving neurological outcome in preterm neonates with hypoxia-induced DWMI.SIGNIFICANCE STATEMENT Diffuse white matter injury (DWMI) caused by hypoxia is a leading cause of neurological deficits following premature birth. An increased understanding of the pathogenesis of this disease is critical. The integrated stress response (ISR) is activated by hypoxia and protects oligodendrocyte lineage cells in other disease models. This has led to an interest in the potential role of the ISR in DWMI. Here we examine the ISR in hypoxia-induced DWMI and show that while the ISR protects oligodendrocyte lineage cells from hypoxia in vitro, genetic inhibition or enhancement of the ISR has no effect on hypoxia-induced DWMI in vivo, suggesting that the ISR does not play a major role in and is not a likely therapeutic target for DWMI.
Collapse
Affiliation(s)
- Benjamin L Clayton
- Department of Neurology, The University of Chicago Center for Peripheral Neuropathy, The University of Chicago, Chicago, Illinois 60637, and
| | - Aaron Huang
- Department of Neurology, The University of Chicago Center for Peripheral Neuropathy, The University of Chicago, Chicago, Illinois 60637, and
| | - Rejani B Kunjamma
- Department of Neurology, The University of Chicago Center for Peripheral Neuropathy, The University of Chicago, Chicago, Illinois 60637, and
| | - Ani Solanki
- Animal Resource Center, The University of Chicago, Chicago, Illinois 60637
| | - Brian Popko
- Department of Neurology, The University of Chicago Center for Peripheral Neuropathy, The University of Chicago, Chicago, Illinois 60637, and
| |
Collapse
|
46
|
Affeldt BM, Obenaus A, Chan J, Pardo AC. Region specific oligodendrocyte transcription factor expression in a model of neonatal hypoxic injury. Int J Dev Neurosci 2017; 61:1-11. [DOI: 10.1016/j.ijdevneu.2017.05.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 05/11/2017] [Accepted: 05/17/2017] [Indexed: 10/19/2022] Open
Affiliation(s)
- Bethann M. Affeldt
- Department of PediatricsLoma Linda University11175 Campus St., Coleman Pavilion Room A1109Loma LindaCA92354USA
| | - Andre Obenaus
- Department of PediatricsLoma Linda University11175 Campus St., Coleman Pavilion Room A1109Loma LindaCA92354USA
- Cell, Molecular and Developmental Biology ProgramUniversity of CaliforniaRiverside, 1140 Bachelor HallRiversideCA92521USA
| | - Jonathan Chan
- Department of PediatricsLoma Linda University11175 Campus St., Coleman Pavilion Room A1109Loma LindaCA92354USA
| | - Andrea C. Pardo
- Department of PediatricsLoma Linda University11175 Campus St., Coleman Pavilion Room A1109Loma LindaCA92354USA
| |
Collapse
|
47
|
Kim HN, Pak ME, Shin MJ, Kim SY, Shin YB, Yun YJ, Shin HK, Choi BT. Comparative analysis of the beneficial effects of treadmill training and electroacupuncture in a rat model of neonatal hypoxia-ischemia. Int J Mol Med 2017; 39:1393-1402. [PMID: 28487967 PMCID: PMC5428954 DOI: 10.3892/ijmm.2017.2970] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 04/19/2017] [Indexed: 12/14/2022] Open
Abstract
In the present study, we investigated whether treadmill training and electroacupuncture (EA) have autonomous or synergistic beneficial effects on deficits caused by neonatal hypoxia-ischemia in Sprague-Dawley rats. For this purpose, rats subjected to hypoxia-ischemia underwent treadmill training and EA stimulation from 4 to 8 weeks of age. Conventional EA (CEA) and scalp EA (SEA) were delivered by electrical stimulation (2 Hz, 1 mA) at traditional acupoints and at the scalp to the primary motor area, respectively. In the behavioral examination, markedly improved performances in the rotarod test were observed in the rats that underwent treadmill exercise, and in the rats that underwent treadmill exercise and CEA compared to the untreated rats subjected to hypoxia-ischemia. An improvement was also observed in the passive avoidance test in the rats that underwent treadmill training and EA. As shown by western blot analysis, the expression levels of neuronal nuclei (NeuN), 2′,3′-cyclic-nucleotide 3′-phosphodiesterase and myelin basic protein (MBP) exhibited a significant decrease in the contralateral subventricular zone (SVZ) of the rats subjected to hypoxia-ischemia compared to the controls; however, these expression levels increased following treadmill exercise and EA stimulation. As shown by immunohistochemical analyses, the thickness of the corpus callosum and the integrated optical density (IOD) of MBP were significantly increased in the rats subjected to treadmill exercise and EA compared to the untreated rats subjected to hypoxiaa-ischemia. The synergistic effects of treadmill training and EA were also observed in the protein levels and IOD of MBP. A marked increase in the number of bromodeoxyuridine (BrdU)- and BrdU/NeuN-positive cells in the contralateral SVZ was also observed in the rats that underwent treadmill training and EA; the number of BrdU-positive cells was synergistically affected by treadmill training and EA. These results suggest that treadmill training and EA stimulation contribute to the enhancement of behavioral recovery following hypoxia-ischemia via the upregulation of myelin components and neurogenesis. Thus, treatment with EA stimulation, as well as treadmill training offers another treatment option to promote functional recovery in cerebral palsy.
Collapse
Affiliation(s)
- Ha Neui Kim
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan, Gyeongsangnam‑do 50612, Republic of Korea
| | - Malk Eun Pak
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan, Gyeongsangnam‑do 50612, Republic of Korea
| | - Myung Jun Shin
- Department of Rehabilitation Medicine, School of Medicine, Pusan National University, Busan 49241, Republic of Korea
| | - Soo Yeon Kim
- Department of Rehabilitation Medicine, Pusan National University, Yangsan Hospital, Yangsan, Gyeongsangnam-do 50612, Republic of Korea
| | - Yong Beom Shin
- Department of Rehabilitation Medicine, School of Medicine, Pusan National University, Busan 49241, Republic of Korea
| | - Young Ju Yun
- Department of Integrative Medicine, School of Korean Medicine, Pusan National University, Yangsan, Gyeongsangnam-do 50612, Republic of Korea
| | - Hwa Kyoung Shin
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan, Gyeongsangnam‑do 50612, Republic of Korea
| | - Byung Tae Choi
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan, Gyeongsangnam‑do 50612, Republic of Korea
| |
Collapse
|
48
|
Morton PD, Ishibashi N, Jonas RA. Neurodevelopmental Abnormalities and Congenital Heart Disease: Insights Into Altered Brain Maturation. Circ Res 2017; 120:960-977. [PMID: 28302742 PMCID: PMC5409515 DOI: 10.1161/circresaha.116.309048] [Citation(s) in RCA: 115] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 12/14/2016] [Accepted: 12/15/2016] [Indexed: 01/14/2023]
Abstract
In the past 2 decades, it has become evident that individuals born with congenital heart disease (CHD) are at risk of developing life-long neurological deficits. Multifactorial risk factors contributing to neurodevelopmental abnormalities associated with CHD have been identified; however, the underlying causes remain largely unknown, and efforts to address this issue have only recently begun. There has been a dramatic shift in focus from newly acquired brain injuries associated with corrective and palliative heart surgery to antenatal and preoperative factors governing altered brain maturation in CHD. In this review, we describe key time windows of development during which the immature brain is vulnerable to injury. Special emphasis is placed on the dynamic nature of cellular events and how CHD may adversely impact the cellular units and networks necessary for proper cognitive and motor function. In addition, we describe current gaps in knowledge and offer perspectives about what can be done to improve our understanding of neurological deficits in CHD. Ultimately, a multidisciplinary approach will be essential to prevent or improve adverse neurodevelopmental outcomes in individuals surviving CHD.
Collapse
Affiliation(s)
- Paul D Morton
- From the Center for Neuroscience Research and Children's National Heart Institute, Children's National Health System, Washington, DC
| | - Nobuyuki Ishibashi
- From the Center for Neuroscience Research and Children's National Heart Institute, Children's National Health System, Washington, DC.
| | - Richard A Jonas
- From the Center for Neuroscience Research and Children's National Heart Institute, Children's National Health System, Washington, DC.
| |
Collapse
|
49
|
Chamberlain KA, Chapey KS, Nanescu SE, Huang JK. Creatine Enhances Mitochondrial-Mediated Oligodendrocyte Survival After Demyelinating Injury. J Neurosci 2017; 37:1479-1492. [PMID: 28069926 PMCID: PMC5299567 DOI: 10.1523/jneurosci.1941-16.2016] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 12/03/2016] [Accepted: 12/28/2016] [Indexed: 01/11/2023] Open
Abstract
Chronic oligodendrocyte loss, which occurs in the demyelinating disorder multiple sclerosis (MS), contributes to axonal dysfunction and neurodegeneration. Current therapies are able to reduce MS severity, but do not prevent transition into the progressive phase of the disease, which is characterized by chronic neurodegeneration. Therefore, pharmacological compounds that promote oligodendrocyte survival could be beneficial for neuroprotection in MS. Here, we investigated the role of creatine, an organic acid involved in adenosine triphosphate (ATP) buffering, in oligodendrocyte function. We found that creatine increased mitochondrial ATP production directly in oligodendrocyte lineage cell cultures and exerted robust protection on oligodendrocytes by preventing cell death in both naive and lipopolysaccharide-treated mixed glia. Moreover, lysolecithin-mediated demyelination in mice deficient in the creatine-synthesizing enzyme guanidinoacetate-methyltransferase (Gamt) did not affect oligodendrocyte precursor cell recruitment, but resulted in exacerbated apoptosis of regenerated oligodendrocytes in central nervous system (CNS) lesions. Remarkably, creatine administration into Gamt-deficient and wild-type mice with demyelinating injury reduced oligodendrocyte apoptosis, thereby increasing oligodendrocyte density and myelin basic protein staining in CNS lesions. We found that creatine did not affect the recruitment of macrophages/microglia into lesions, suggesting that creatine affects oligodendrocyte survival independently of inflammation. Together, our results demonstrate a novel function for creatine in promoting oligodendrocyte viability during CNS remyelination.SIGNIFICANCE STATEMENT We report that creatine enhances oligodendrocyte mitochondrial function and protects against caspase-dependent oligodendrocyte apoptosis during CNS remyelination. This work has important implications for the development of therapeutic targets for diseases characterized by oligodendrocyte death, including multiple sclerosis.
Collapse
Affiliation(s)
- Kelly A Chamberlain
- Department of Biology and
- Interdisciplinary Program in Neuroscience, Georgetown University, Washington, District of Columbia 20057
| | | | | | - Jeffrey K Huang
- Department of Biology and
- Interdisciplinary Program in Neuroscience, Georgetown University, Washington, District of Columbia 20057
| |
Collapse
|
50
|
Sirt1 regulates glial progenitor proliferation and regeneration in white matter after neonatal brain injury. Nat Commun 2016; 7:13866. [PMID: 27991597 PMCID: PMC5187440 DOI: 10.1038/ncomms13866] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 11/07/2016] [Indexed: 11/15/2022] Open
Abstract
Regenerative processes in brain pathologies require the production of distinct neural cell populations from endogenous progenitor cells. We have previously demonstrated that oligodendrocyte progenitor cell (OPC) proliferation is crucial for oligodendrocyte (OL) regeneration in a mouse model of neonatal hypoxia (HX) that reproduces diffuse white matter injury (DWMI) of premature infants. Here we identify the histone deacetylase Sirt1 as a Cdk2 regulator in OPC proliferation and response to HX. HX enhances Sirt1 and Sirt1/Cdk2 complex formation through HIF1α activation. Sirt1 deacetylates retinoblastoma (Rb) in the Rb/E2F1 complex, leading to dissociation of E2F1 and enhanced OPC proliferation. Sirt1 knockdown in culture and its targeted ablation in vivo suppresses basal and HX-induced OPC proliferation. Inhibition of Sirt1 also promotes OPC differentiation after HX. Our results indicate that Sirt1 is an essential regulator of OPC proliferation and OL regeneration after neonatal brain injury. Therefore, enhancing Sirt1 activity may promote OL recovery after DWMI.
Oligodendrocyte progenitor cell (OPC) proliferation is crucial for regeneration after hypoxic lesions in mice, a model of diffuse white matter injury of premature infants. Here, the authors show that the histone deacetylase Sirt1 is a Cdk2-dependent mediator of OPC proliferation and OPC response to hypoxia.
Collapse
|