1
|
Dalenogare DP, Souza Monteiro de Araújo D, Landini L, Titiz M, De Siena G, De Logu F, Geppetti P, Nassini R, Trevisan G. Neuropathic-like Nociception and Spinal Cord Neuroinflammation Are Dependent on the TRPA1 Channel in Multiple Sclerosis Models in Mice. Cells 2023; 12:1511. [PMID: 37296632 PMCID: PMC10252670 DOI: 10.3390/cells12111511] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 05/08/2023] [Accepted: 05/26/2023] [Indexed: 06/12/2023] Open
Abstract
Background: Transient receptor potential ankyrin 1 (TRPA1) activation is implicated in neuropathic pain-like symptoms. However, whether TRPA1 is solely implicated in pain-signaling or contributes to neuroinflammation in multiple sclerosis (MS) is unknown. Here, we evaluated the TRPA1 role in neuroinflammation underlying pain-like symptoms using two different models of MS. Methods: Using a myelin antigen, Trpa1+/+ or Trpa1-/- female mice developed relapsing-remitting experimental autoimmune encephalomyelitis (RR-EAE) (Quil A as adjuvant) or progressive experimental autoimmune encephalomyelitis (PMS)-EAE (complete Freund's adjuvant). The locomotor performance, clinical scores, mechanical/cold allodynia, and neuroinflammatory MS markers were evaluated. Results: Mechanical and cold allodynia detected in RR-EAE, or PMS-EAE Trpa1+/+ mice, were not observed in Trpa1-/- mice. The increased number of cells labeled for ionized calcium-binding adapter molecule 1 (Iba1) or glial fibrillary acidic protein (GFAP), two neuroinflammatory markers in the spinal cord observed in both RR-EAE or PMS-EAE Trpa1+/+ mice, was reduced in Trpa1-/- mice. By Olig2 marker and luxol fast blue staining, prevention of the demyelinating process in Trpa1-/- induced mice was also detected. Conclusions: Present results indicate that the proalgesic role of TRPA1 in EAE mouse models is primarily mediated by its ability to promote spinal neuroinflammation and further strengthen the channel inhibition to treat neuropathic pain in MS.
Collapse
Affiliation(s)
- Diéssica Padilha Dalenogare
- Graduated Program in Pharmacology, Federal University of Santa Maria (UFSM), Santa Maria 97105-900, RS, Brazil;
| | - Daniel Souza Monteiro de Araújo
- Clinical Pharmacology Unit, Department of Health Sciences, University of Florence, 50139 Florence, Italy; (D.S.M.d.A.); (L.L.); (M.T.); (G.D.S.); (F.D.L.); (P.G.); (R.N.)
| | - Lorenzo Landini
- Clinical Pharmacology Unit, Department of Health Sciences, University of Florence, 50139 Florence, Italy; (D.S.M.d.A.); (L.L.); (M.T.); (G.D.S.); (F.D.L.); (P.G.); (R.N.)
| | - Mustafa Titiz
- Clinical Pharmacology Unit, Department of Health Sciences, University of Florence, 50139 Florence, Italy; (D.S.M.d.A.); (L.L.); (M.T.); (G.D.S.); (F.D.L.); (P.G.); (R.N.)
| | - Gaetano De Siena
- Clinical Pharmacology Unit, Department of Health Sciences, University of Florence, 50139 Florence, Italy; (D.S.M.d.A.); (L.L.); (M.T.); (G.D.S.); (F.D.L.); (P.G.); (R.N.)
| | - Francesco De Logu
- Clinical Pharmacology Unit, Department of Health Sciences, University of Florence, 50139 Florence, Italy; (D.S.M.d.A.); (L.L.); (M.T.); (G.D.S.); (F.D.L.); (P.G.); (R.N.)
| | - Pierangelo Geppetti
- Clinical Pharmacology Unit, Department of Health Sciences, University of Florence, 50139 Florence, Italy; (D.S.M.d.A.); (L.L.); (M.T.); (G.D.S.); (F.D.L.); (P.G.); (R.N.)
| | - Romina Nassini
- Clinical Pharmacology Unit, Department of Health Sciences, University of Florence, 50139 Florence, Italy; (D.S.M.d.A.); (L.L.); (M.T.); (G.D.S.); (F.D.L.); (P.G.); (R.N.)
| | - Gabriela Trevisan
- Graduated Program in Pharmacology, Federal University of Santa Maria (UFSM), Santa Maria 97105-900, RS, Brazil;
| |
Collapse
|
2
|
Savchuk S, Monje M. Mini-Review: Aplastic Myelin Following Chemotherapy. Neurosci Lett 2022; 790:136861. [PMID: 36055447 DOI: 10.1016/j.neulet.2022.136861] [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: 12/09/2021] [Revised: 05/12/2022] [Accepted: 08/26/2022] [Indexed: 11/16/2022]
Abstract
The contribution of chemotherapy to improved outcomes for cancer patients is unquestionable. Yet as its applications broaden, so do the concerns for the long-term implications of chemotherapy on the health of cancer survivors, with chemotherapy-related cognitive impairment as a cause for particular urgency. In this mini review, we explore myelin aplasticity following chemotherapy, discussing the role of myelin plasticity in healthy cognition and failure of myelin plasticity chiefly due microenvironmental aberrations in chemotherapy-related cognitive impairment. Possible therapeutic strategies to mitigate chemotherapy-induced myelin dysfunction are also discussed.
Collapse
Affiliation(s)
- Solomiia Savchuk
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, 94305, USA
| | - Michelle Monje
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, 94305, USA; Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, 94305, USA; Department of Neurosurgery, Stanford University, Stanford, CA, 94305, USA; Department of Pathology, Stanford University, Stanford, CA, 94305, USA; Department of Pediatrics, Stanford University, Stanford, CA, 94305, USA; Howard Hughes Medical Institute, Stanford University, Stanford, CA, 94305, USA.
| |
Collapse
|
3
|
Yuan H, Zhang B, Ma J, Zhang Y, Tuo Y, Li X. Analysis of gene expression profiles in two spinal cord injury models. Eur J Med Res 2022; 27:156. [PMID: 35999613 PMCID: PMC9400253 DOI: 10.1186/s40001-022-00785-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 08/04/2022] [Indexed: 11/25/2022] Open
Abstract
Objectives To analyze the changes of gene expression at different timepoints after spinal cord injury (SCI) with tenth segment thoracic injury. Methods Two SCI models, the complete paraplegia (H) and Allen’s strike (D) methods were applied to induce SCI in rats, and transcriptome sequencing was performed 1, 3, 7, 14, 56, and 70 days after SCI, respectively. Principal component analysis, differentially expressed gene analysis, and hierarchical clustering analysis were applied to analyze the differentially expressed genes (DEGs). Gene Ontology GO enrichment analysis, Kyoto Encyclopedia of Genes and Genomes enrichment analysis, and Gene Set Enrichment Analysis revealed the pathway of gene enrichment. Results There were 1,907, 3,120, 3,728, 978, 2,319, and 3,798 DEGs in the complete paraplegia group and 2,380, 878, 1,543, 6,040, 1,945, and 3,850 DEGs in the Allen’s strike method group and after SCI at 1, 3, 7, 14, 56, and 70 days, respectively. The transcriptome contours of D1, H1, D3, and H14 were clustered with C; the H56, D56, H70, and D70 transcriptome contours were similar and clustered together. H3, D7, and H7 were clustered together, and D14 was clustered separately. The transcriptome differences of the two SCI models were mainly concentrated during the first 2 weeks after SCI. The DEGs after SCI in the complete paraplegia group were more concentrated. Most of the early transcriptional regulation stabilized within 2 weeks after injury. Conclusions There were DEGs between the two SCI models. Through the gene changes and pathway enrichment of the entire time period after SCI, the molecular mechanism of SCI repair was revealed in depth, which provided a reference for SCI treatment in the future.
Collapse
Affiliation(s)
- Haifeng Yuan
- Department of Spinal Orthopedics, General Hospital of Ningxia Medical University, No. 804 Shengli Street, Xingqing District, Yinchuan, 750004, China
| | - Bi Zhang
- Department of Anesthesia, Ningbo Medical Center Li Huili Hospital, Ningbo, 315046, China
| | - Junchi Ma
- Department of Orthopaedics, Affiliated Hospital of Gansu College of Traditional Chinese Medicine, Lanzhou, 730099, China
| | - Yufei Zhang
- The third department of spine, Baoji Hospital of Traditional Chinese Medicine, Baoji, 721001, China
| | - Yifan Tuo
- Clinical Medicine, Ningxia Medical University, Yinchuan, 750004, China
| | - Xusheng Li
- Department of Spinal Orthopedics, General Hospital of Ningxia Medical University, No. 804 Shengli Street, Xingqing District, Yinchuan, 750004, China.
| |
Collapse
|
4
|
Dermitzakis I, Manthou ME, Meditskou S, Miliaras D, Kesidou E, Boziki M, Petratos S, Grigoriadis N, Theotokis P. Developmental Cues and Molecular Drivers in Myelinogenesis: Revisiting Early Life to Re-Evaluate the Integrity of CNS Myelin. Curr Issues Mol Biol 2022; 44:3208-3237. [PMID: 35877446 PMCID: PMC9324160 DOI: 10.3390/cimb44070222] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/14/2022] [Accepted: 07/17/2022] [Indexed: 02/07/2023] Open
Abstract
The mammalian central nervous system (CNS) coordinates its communication through saltatory conduction, facilitated by myelin-forming oligodendrocytes (OLs). Despite the fact that neurogenesis from stem cell niches has caught the majority of attention in recent years, oligodendrogenesis and, more specifically, the molecular underpinnings behind OL-dependent myelinogenesis, remain largely unknown. In this comprehensive review, we determine the developmental cues and molecular drivers which regulate normal myelination both at the prenatal and postnatal periods. We have indexed the individual stages of myelinogenesis sequentially; from the initiation of oligodendrocyte precursor cells, including migration and proliferation, to first contact with the axon that enlists positive and negative regulators for myelination, until the ultimate maintenance of the axon ensheathment and myelin growth. Here, we highlight multiple developmental pathways that are key to successful myelin formation and define the molecular pathways that can potentially be targets for pharmacological interventions in a variety of neurological disorders that exhibit demyelination.
Collapse
Affiliation(s)
- Iasonas Dermitzakis
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (I.D.); (M.E.M.); (S.M.); (D.M.)
| | - Maria Eleni Manthou
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (I.D.); (M.E.M.); (S.M.); (D.M.)
| | - Soultana Meditskou
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (I.D.); (M.E.M.); (S.M.); (D.M.)
| | - Dimosthenis Miliaras
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (I.D.); (M.E.M.); (S.M.); (D.M.)
| | - Evangelia Kesidou
- Laboratory of Experimental Neurology and Neuroimmunology, Second Department of Neurology, AHEPA University Hospital, 54621 Thessaloniki, Greece; (E.K.); (M.B.); (N.G.)
| | - Marina Boziki
- Laboratory of Experimental Neurology and Neuroimmunology, Second Department of Neurology, AHEPA University Hospital, 54621 Thessaloniki, Greece; (E.K.); (M.B.); (N.G.)
| | - Steven Petratos
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC 3004, Australia;
| | - Nikolaos Grigoriadis
- Laboratory of Experimental Neurology and Neuroimmunology, Second Department of Neurology, AHEPA University Hospital, 54621 Thessaloniki, Greece; (E.K.); (M.B.); (N.G.)
| | - Paschalis Theotokis
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (I.D.); (M.E.M.); (S.M.); (D.M.)
- Laboratory of Experimental Neurology and Neuroimmunology, Second Department of Neurology, AHEPA University Hospital, 54621 Thessaloniki, Greece; (E.K.); (M.B.); (N.G.)
- Correspondence:
| |
Collapse
|
5
|
Safaei HA, Eftekhari SM, Aliomrani M. Analysis of platelet-derived growth factor receptor A and oligodendrocyte transcription factor 2 markers following Hydroxychloroquine administration in animal induced multiple sclerosis model. Metab Brain Dis 2021; 36:2101-2110. [PMID: 34342813 DOI: 10.1007/s11011-021-00802-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 07/22/2021] [Indexed: 12/19/2022]
Abstract
It has been shown that following demyelination, Oligodendrocyte Progenitor Cells (OPCs) migrate to the lesion site and begin to proliferate, and differentiate. This study aimed to investigate the effects of Hydroxychloroquine (HCQ) on the expression of OLIG-2 and PDGFR-α markers during the myelination process. C57BL/6 mice were fed cuprizone pellets for 5 weeks to induce demyelination and return to a normal diet for 1 week to stimulate remyelination. During the Phase I all of the animals except CPZ and Vehicle groups were exposed to HCQ (2.5, 10, and 100 mg/kg) via drinking water. At the end of the study, animals were euthanized, perfused and the brain samples were assessed for myelination and immunohistochemistry evaluation. What is remarkable is the high rate of Olig2 + cells in the groups treated with 10 and 100 mg/kg HCQ in the demyelination phase and its decreasing trend in the remyelination phase. However, there was no significant difference between groups during phase I and Phase II based on the percentage of olig-2+/total cells in the corpus callosum region. The number of PDGFR-α+ cells in the group treated with 10 mg/kg HCQ was significant in the first phase (p value < 0.05). Considering that the 100 mg/kg HCQ group had the highest level of PDGFR-α as well as the highest level of myelin repair in LFB staining, it could be inferred that it was the most effective dose in inducing proliferation and migration of OPCs.
Collapse
Affiliation(s)
- Hajar Amin Safaei
- School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences and Health Services, Isfahan, Iran
| | | | - Mehdi Aliomrani
- Department of Toxicology and Pharmacology, Isfahan Pharmaceutical Science Research Center, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, I. R. of Iran.
| |
Collapse
|
6
|
The Effects of the Olig Family on the Regulation of Spinal Cord Development and Regeneration. Neurochem Res 2021; 46:2776-2782. [PMID: 34228233 DOI: 10.1007/s11064-021-03383-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 06/15/2021] [Accepted: 06/17/2021] [Indexed: 12/22/2022]
Abstract
Neurons and glial cells in the central nervous system (CNS) are generated from neuroepithelial cells in the ventricular zone that surrounds the embryonic neural tube. The proliferation and distinct differentiation of neural precursors occurs at certain stages and are regulated by a series of transcription factors leading to the generation of neuronal and glial cell subtypes. In this manuscript, we review the effects of the Olig family, namely, members Olig1, Olig2 and Olig3, on the distinct differentiation of glial and neuronal cells in the developing spinal cord and injured neural tissue.
Collapse
|
7
|
Wang X, Su Y, Li T, Yu G, Wang Y, Chen X, Yin C, Tang Z, Yi C, Xiao L, Niu J. Quetiapine promotes oligodendroglial process outgrowth and membrane expansion by orchestrating the effects of Olig1. Glia 2021; 69:1709-1722. [PMID: 33660902 DOI: 10.1002/glia.23986] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 02/18/2021] [Accepted: 02/18/2021] [Indexed: 01/09/2023]
Abstract
Oligodendroglial lineage cells go through a series of morphological changes before myelination. Prior to myelination, cell processes and membrane structures enlarge by approximately 7,000 times, which is required to support axonal wrapping and myelin segment formation. Failure of these processes leads to maldevelopment and impaired myelination. Quetiapine, an atypical antipsychotic drug, was proved to promote oligodendroglial differentiation and (re)myelination, pending detailed effects and regulatory mechanism. In this study, we showed that quetiapine promotes morphological maturation of oligodendroglial lineage cells and myelin segment formation, and a short-term quetiapine treatment is sufficient to induce these changes. To uncover the underlying mechanism, we examined the effect of quetiapine on the Oligodendrocyte transcription factor 1 (Olig1). We found that quetiapine upregulates Olig1 expression level and promotes nuclear Olig1 translocation to the cytosol, where it functions not as a transcription modulator, but in a way that highly correlates with oligodendrocyte morphological transformation. In addition, quetiapine treatment reverses the negative regulatory effect of the Olig1-regulated G protein-coupled receptor 17 (GPR17) on oligodendroglial morphological maturation. Our results demonstrate that quetiapine enhances oligodendroglial differentiation and myelination by promoting cell morphological transformation. This would shed light on the orchestration of oligodendroglia developmental mechanisms, and provides new targets for further therapeutic research.
Collapse
Affiliation(s)
- Xiaorui Wang
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing, China
| | - Yixun Su
- Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Tao Li
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing, China
| | - Guangdan Yu
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing, China
| | - Yuxin Wang
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing, China
| | - Xiaoying Chen
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing, China
| | - Chenrui Yin
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing, China
| | - Ziqin Tang
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing, China
| | - Chenju Yi
- Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Lan Xiao
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing, China
| | - Jianqin Niu
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing, China
| |
Collapse
|
8
|
Khalilian B, Madadi S, Fattahi N, Abouhamzeh B. Coenzyme Q10 enhances remyelination and regulate inflammation effects of cuprizone in corpus callosum of chronic model of multiple sclerosis. J Mol Histol 2021; 52:125-134. [PMID: 33245472 DOI: 10.1007/s10735-020-09929-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 11/17/2020] [Indexed: 12/30/2022]
Abstract
Multiple Sclerosis (MS) is a chronic, progressive demyelinating disease of the central nervous system that causes the most disability in young people, besides trauma. Coenzyme Q10 (CoQ10)-also known as ubiquinone-is an endogenous lipid-soluble antioxidant in the mitochondrial oxidative respiratory chain which can reduce oxidative stress and inflammation, the processes associated with demyelination in MS. Cuprizone (CPZ) intoxication is a well-established model of inducing MS, best for studying demyelination-remyelination. In this study, we examined for the first time the role of CoQ10 in preventing demyelination and induction of remyelination in the chronic CPZ model of MS. 40 male mice were divided into four groups. 3 group chewed CPZ-containing food for 12 weeks to induce MS. After 4 weeks, one group were treated with CoQ10 (150 mg/kg/day) by daily gavage until the end of the experiment, while CPZ poisoning continued. At the end of 12 weeks, tail suspension test (TST) and open field test (OFT) was taken and animals were sacrificed to assess myelin basic protein (MBP), oligodendrocyte transcription factor-1 (Olig1), tumor necrosis factor-α (TNF-α) and interleukin 6 (IL-6) by real-time polymerase chain reaction (real-time PCR) and total antioxidant capacity (TAC) and superoxide dismutase (SOD) by Elisa test. Luxol fast blue (LFB) staining was used to evaluate histological changes. CoQ10 administration promoted remyelination in histological findings. MBP and Olig-1 expression were increased significantly in CoQ10 treated group compare to the CPZ-intoxicated group. CoQ10 treatment alleviated stress oxidative status induced by CPZ and dramatically suppress inflammatory biomarkers. CPZ ingestion made no significant difference between normal control group and the CPZ-intoxicated group in TST and OFT. CoQ10 can enhance remyelination in the CPZ model and potentially might have same effects in MS patients.
Collapse
Affiliation(s)
- Behnam Khalilian
- Department of Anatomical Sciences, Faculty of Medicine, AJA University of Medical Sciences, 1411718541, Tehran, Iran
| | - Soheila Madadi
- Department of Anatomy, Faculty of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Nima Fattahi
- Non-communicable Diseases Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Beheshteh Abouhamzeh
- Department of Anatomical Sciences, Faculty of Medicine, AJA University of Medical Sciences, 1411718541, Tehran, Iran.
| |
Collapse
|
9
|
Pro-Inflammatory Signaling Upregulates a Neurotoxic Conotoxin-Like Protein Encrypted Within Human Endogenous Retrovirus-K. Cells 2020; 9:cells9071584. [PMID: 32629888 PMCID: PMC7407490 DOI: 10.3390/cells9071584] [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: 05/30/2020] [Revised: 06/20/2020] [Accepted: 06/25/2020] [Indexed: 02/06/2023] Open
Abstract
Motor neuron degeneration and spinal cord demyelination are hallmark pathological events in Amyotrophic Lateral Sclerosis (ALS). Endogenous retrovirus-K (ERVK) expression has an established association with ALS neuropathology, with murine modeling pointing to a role for the ERVK envelope (env) gene in disease processes. Here, we describe a novel viral protein cryptically encoded within the ERVK env transcript, which resembles two distinct cysteine-rich neurotoxic proteins: conotoxin proteins found in marine snails and the Human Immunodeficiency Virus (HIV) Tat protein. Consistent with Nuclear factor-kappa B (NF-κB)-induced retrotransposon expression, the ERVK conotoxin-like protein (CTXLP) is induced by inflammatory signaling. CTXLP is found in the nucleus, impacting innate immune gene expression and NF-κB p65 activity. Using human autopsy specimens from patients with ALS, we further showcase CTXLP expression in degenerating motor cortex and spinal cord tissues, concomitant with inflammation linked pathways, including enhancement of necroptosis marker mixed lineage kinase domain-like (MLKL) protein and oligodendrocyte maturation/myelination inhibitor Nogo-A. These findings identify CTXLP as a novel ERVK protein product, which may act as an effector in ALS neuropathology.
Collapse
|
10
|
Kumar S, Fritz Z, Sulakhiya K, Theis T, Berthiaume F. Transcriptional Factors and Protein Biomarkers as Target Therapeutics in Traumatic Spinal Cord and Brain Injury. Curr Neuropharmacol 2020; 18:1092-1105. [PMID: 32442086 PMCID: PMC7709155 DOI: 10.2174/1570159x18666200522203542] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 03/19/2020] [Accepted: 05/07/2020] [Indexed: 12/04/2022] Open
Abstract
Traumatic injury to the spinal cord (SCI) and brain (TBI) are serious health problems and affect many people every year throughout the world. These devastating injuries are affecting not only patients but also their families socially as well as financially. SCI and TBI lead to neurological dysfunction besides continuous inflammation, ischemia, and necrosis followed by progressive neurodegeneration. There are well-established changes in several other processes such as gene expression as well as protein levels that are the important key factors to control the progression of these diseases. We are not yet able to collect enough knowledge on the underlying mechanisms leading to the altered gene expression profiles and protein levels in SCI and TBI. Cell loss is hastened by the induction or imbalance of pro- or anti-inflammatory expression profiles and transcription factors for cell survival after or during trauma. There is a sequence of events of dysregulation of these factors from early to late stages of trauma that opens a therapeutic window for new interventions to prevent/restrict the progression of these diseases. There has been increasing interest in the modulation of these factors for improving the patient’s quality of life by targeting both SCI and TBI. Here, we review some of the recent transcriptional factors and protein biomarkers that have been developed and discovered in the last decade in the context of targeted therapeutics for SCI and TBI patients.
Collapse
Affiliation(s)
- Suneel Kumar
- Department of Biomedical Engineering, The State University of New Jersey, Piscataway, New Jersey, USA
| | - Zachary Fritz
- Department of Biomedical Engineering, The State University of New Jersey, Piscataway, New Jersey, USA
| | - Kunjbihari Sulakhiya
- Department of Pharmacy, Indira Gandhi National Tribal University (IGNTU), Amarkantak, India
| | - Thomas Theis
- W. M. Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers, The
State University of New Jersey, Piscataway, New Jersey, USA
| | - Francois Berthiaume
- Department of Biomedical Engineering, The State University of New Jersey, Piscataway, New Jersey, USA
| |
Collapse
|
11
|
Dziedzic A, Miller E, Saluk-Bijak J, Bijak M. The GPR17 Receptor-A Promising Goal for Therapy and a Potential Marker of the Neurodegenerative Process in Multiple Sclerosis. Int J Mol Sci 2020; 21:ijms21051852. [PMID: 32182666 PMCID: PMC7084627 DOI: 10.3390/ijms21051852] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 02/29/2020] [Accepted: 03/06/2020] [Indexed: 12/28/2022] Open
Abstract
One of the most important goals in the treatment of demyelinating diseases such as multiple sclerosis (MS) is, in addition to immunomodulation, reconstruction of the lost myelin sheath. The modulator of the central nervous system myelination is the metabotropic receptor coupled to the G-protein: GPR17. GPR17 receptors are considered to be sensors of local damage to the myelin sheath, and play a role in the reconstruction and repair of demyelinating plaques caused by ongoing inflammatory processes. GPR17 receptors are present on nerve cells and precursor oligodendrocyte cells. Under physiological conditions, they are responsible for the differentiation and subsequent maturation of oligodendrocytes, while under pathological conditions (during damage to nerve cells), their expression increases to become mediators in the demyelinating processes. Moreover, they are essential not only in both the processes of inducing damage and the death of neurons, but also in the local repair of the damaged myelin sheath. Therefore, GPR17 receptors may be recognized as the potential goal in creating innovative therapies for the treatment of the neurodegenerative process in MS, based on the acceleration of the remyelination processes. This review examines the role of GRP17 in pathomechanisms of MS development.
Collapse
Affiliation(s)
- Angela Dziedzic
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland; (A.D.); (J.S.-B.)
| | - Elzbieta Miller
- Department of Neurological Rehabilitation, Medical University of Lodz, Milionowa 14, 93-113 Lodz, Poland;
| | - Joanna Saluk-Bijak
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland; (A.D.); (J.S.-B.)
| | - Michal Bijak
- Biohazard Prevention Centre, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland
- Correspondence: ; Tel.: +48-42-635-4336
| |
Collapse
|
12
|
Bisphenol A triggers axonal injury and myelin degeneration with concomitant neurobehavioral toxicity in C57BL/6J male mice. Toxicology 2019; 428:152299. [PMID: 31574244 DOI: 10.1016/j.tox.2019.152299] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 09/10/2019] [Accepted: 09/25/2019] [Indexed: 12/20/2022]
Abstract
Bisphenol A (BPA) is a ubiquitously distributed endocrine disrupting chemical (EDC). BPA exposure in humans has been a matter of concern due to its increased application in the products of day to day use. BPA has been reported to cause toxicity in almost all the vital organ systems even at a very low dose levels. It crosses the blood brain barrier and causes neurotoxicity. We studied the effect of BPA on the cerebral cortex of C57BL/6J mice and examined whether BPA exposure alters the expression of axonal and myelin structural proteins. Male mice were dosed orally to 40 μg and 400 μg BPA/kg body weight for 60 days. BPA exposure resulted in memory loss, muscle coordination deficits and allodynia. BPA exposure also caused degeneration of immature and mature oligodendrocytes as evaluated by decreased mRNA levels of 2',3'-cyclic nucleotide 3' phosphodiesterase (CNPase), nestin, myelin basic protein (MBP) and myelin-associated glycoprotein-1 (MAG-1) genes revealing myelin related pathology. It was observed that subchronic BPA exposure caused neuroinflammation through deregulation of inflammatory cytokines mRNA and protein expression which further resulted into neurotoxicity through axonal as well as myelin degeneration in the brain. BPA also caused increased oxidative stress in the brain. Our study indicates long-term subchronic low dose exposure to BPA has the potential to cause axonal degeneration and demyelination in the oligodendrocytes and neurons which may have implications in neurological and neuropsychological disorders including multiple sclerosis (MS), neuromyelitis optica and others.
Collapse
|
13
|
Initial findings of striatum tripartite model in OCD brain samples based on transcriptome analysis. Sci Rep 2019; 9:3086. [PMID: 30816141 PMCID: PMC6395771 DOI: 10.1038/s41598-019-38965-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 12/17/2018] [Indexed: 11/22/2022] Open
Abstract
Obsessive-compulsive disorder (OCD) is a psychiatric disorder characterized by obsessions and/or compulsions. Different striatal subregions belonging to the cortico-striato-thalamic circuitry (CSTC) play an important role in the pathophysiology of OCD. The transcriptomes of 3 separate striatal areas (putamen (PT), caudate nucleus (CN) and accumbens nucleus (NAC)) from postmortem brain tissue were compared between 6 OCD and 8 control cases. In addition to network connectivity deregulation, different biological processes are specific to each striatum region according to the tripartite model of the striatum and contribute in various ways to OCD pathophysiology. Specifically, regulation of neurotransmitter levels and presynaptic processes involved in chemical synaptic transmission were shared between NAC and PT. The Gene Ontology terms cellular response to chemical stimulus, response to external stimulus, response to organic substance, regulation of synaptic plasticity, and modulation of synaptic transmission were shared between CN and PT. Most genes harboring common and/or rare variants previously associated with OCD that were differentially expressed or part of a least preserved coexpression module in our study also suggest striatum subregion specificity. At the transcriptional level, our study supports differences in the 3 circuit CSTC model associated with OCD.
Collapse
|
14
|
Schlüter A, Sandoval J, Fourcade S, Díaz-Lagares A, Ruiz M, Casaccia P, Esteller M, Pujol A. Epigenomic signature of adrenoleukodystrophy predicts compromised oligodendrocyte differentiation. Brain Pathol 2018; 28:902-919. [PMID: 29476661 PMCID: PMC6857458 DOI: 10.1111/bpa.12595] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 02/13/2018] [Accepted: 02/16/2018] [Indexed: 12/19/2022] Open
Abstract
Epigenomic changes may either cause disease or modulate its expressivity, adding a layer of complexity to mendelian diseases. X‐linked adrenoleukodystrophy (X‐ALD) is a rare neurometabolic condition exhibiting discordant phenotypes, ranging from a childhood cerebral inflammatory demyelination (cALD) to an adult‐onset mild axonopathy in spinal cords (AMN). The AMN form may occur with superimposed inflammatory brain demyelination (cAMN). All patients harbor loss of function mutations in the ABCD1 peroxisomal transporter of very‐long chain fatty acids. The factors that account for the lack of genotype‐phenotype correlation, even within the same family, remain largely unknown. To gain insight into this matter, here we compared the genome‐wide DNA methylation profiles of morphologically intact frontal white matter areas of children affected by cALD with adult cAMN patients, including male controls in the same age group. We identified a common methylomic signature between the two phenotypes, comprising (i) hypermethylation of genes harboring the H3K27me3 mark at promoter regions, (ii) hypermethylation of genes with major roles in oligodendrocyte differentiation such as MBP, CNP, MOG and PLP1 and (iii) hypomethylation of immune‐associated genes such as IFITM1 and CD59. Moreover, we found increased hypermethylation in CpGs of genes involved in oligodendrocyte differentiation, and also in genes with H3K27me3 marks in their promoter regions in cALD compared with cAMN, correlating with transcriptional and translational changes. Further, using a penalized logistic regression model, we identified the combined methylation levels of SPG20, UNC45A and COL9A3 and also, the combined expression levels of ID4 and MYRF to be good markers capable of discriminating childhood from adult inflammatory phenotypes. We thus propose the hypothesis that an epigenetically controlled, altered transcriptional program may drive an impaired oligodendrocyte differentiation and aberrant immune activation in X‐ALD patients. These results shed light into disease pathomechanisms and uncover putative biomarkers of interest for prognosis and phenotypic stratification.
Collapse
Affiliation(s)
- Agatha Schlüter
- Neurometabolic Diseases Laboratory, Bellvitge Biomedical Research Institute (IDIBELL), 08908 L'Hospitalet de Llobregat, Barcelona, Spain.,Center for Biomedical Research on Rare Diseases (CIBERER), ISCIII, Spain
| | - Juan Sandoval
- Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Catalonia, Spain
| | - Stéphane Fourcade
- Neurometabolic Diseases Laboratory, Bellvitge Biomedical Research Institute (IDIBELL), 08908 L'Hospitalet de Llobregat, Barcelona, Spain.,Center for Biomedical Research on Rare Diseases (CIBERER), ISCIII, Spain
| | - Angel Díaz-Lagares
- Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Catalonia, Spain
| | - Montserrat Ruiz
- Neurometabolic Diseases Laboratory, Bellvitge Biomedical Research Institute (IDIBELL), 08908 L'Hospitalet de Llobregat, Barcelona, Spain.,Center for Biomedical Research on Rare Diseases (CIBERER), ISCIII, Spain
| | - Patrizia Casaccia
- Department of Neuroscience and Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029.,Neuroscience Initiative ASRC CUNY, 85 St Nicholas Terrace, New York, NY 10031
| | - Manel Esteller
- Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Catalonia, Spain.,Physiological Sciences Department, School of Medicine and Health Sciences, University of Barcelona (UB), Catalonia, Spain.,Catalan Institution of Research and Advanced Studies (ICREA), Barcelona, Catalonia, Spain
| | - Aurora Pujol
- Neurometabolic Diseases Laboratory, Bellvitge Biomedical Research Institute (IDIBELL), 08908 L'Hospitalet de Llobregat, Barcelona, Spain.,Center for Biomedical Research on Rare Diseases (CIBERER), ISCIII, Spain.,Catalan Institution of Research and Advanced Studies (ICREA), Barcelona, Catalonia, Spain
| |
Collapse
|
15
|
Impaired oligodendrogenesis and myelination by elevated S100B levels during neurodevelopment. Neuropharmacology 2018; 129:69-83. [DOI: 10.1016/j.neuropharm.2017.11.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 10/22/2017] [Accepted: 11/03/2017] [Indexed: 11/23/2022]
|
16
|
Mizee MR, Poel MVD, Huitinga I. Purification of cells from fresh human brain tissue: primary human glial cells. HANDBOOK OF CLINICAL NEUROLOGY 2018; 150:273-283. [PMID: 29496146 DOI: 10.1016/b978-0-444-63639-3.00019-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
In order to translate the findings obtained from postmortem brain tissue samples to functional biologic mechanisms of central nervous system disease, it will be necessary to understand how these findings affect the different cell populations in the brain. The acute isolation and analysis of pure glial cell populations are common practice in animal models for neurologic diseases, but are not yet regularly applied to human postmortem brain material. The development of novel cell isolation techniques and methods for transcriptomic and proteomic analysis have made it possible to isolate and phenotype primary human cell populations from the central nervous system. The psychiatric program of the Netherlands Brain Bank has considerable experience with the purification of glial cells. This chapter will review the rapid isolation and phenotyping procedures for two major glia cell populations in the human brain, microglia and astrocytes, and will also discuss the potential for biobanking these cells, as well as the possible alternatives to cell isolations. The acute isolation of glial cells without culture-based adherence steps allows the analysis of glial alterations that underlie, or are the result of, disease neuropathology of the donor.
Collapse
Affiliation(s)
- Mark Ronald Mizee
- Neuroimmunology Research Group, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands; Psychiatric Program, Netherlands Brain Bank, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Marlijn van der Poel
- Neuroimmunology Research Group, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Inge Huitinga
- Neuroimmunology Research Group, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands; Psychiatric Program, Netherlands Brain Bank, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands.
| |
Collapse
|
17
|
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: 0.6] [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
|
18
|
Yang T, Zheng Q, Wang S, Fang L, Liu L, Zhao H, Wang L, Fan Y. Effect of catalpol on remyelination through experimental autoimmune encephalomyelitis acting to promote Olig1 and Olig2 expressions in mice. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2017; 17:240. [PMID: 28464811 PMCID: PMC5414219 DOI: 10.1186/s12906-017-1642-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 02/21/2017] [Indexed: 11/30/2022]
Abstract
BACKGROUND Multiple sclerosis (MS) as an autoimmune disorder is a common disease occurring in central nervous system (CNS) and the remyelination plays a pivotal role in the alleviating neurological impairment in the MS. Catalpol, an effective component extracted from the Chinese herb Radix Rehmanniae, which has been proved protective in cerebral diseases. METHODS To determine the protective effects and mechanisms of Catalpol on MS, the mice with experimental autoimmune encephalomyelitis (EAE) were induced by myelin oligodendrocyte glycoprotein (MOG) 35-55, as a model for human MS. Th17 cells were counted by flow cytometric (FCM). The expressions of nerve-glial antigen (NG) 2 and myelin basic protein (MBP) were measured by immunohistochemical staining. Olig1+ and Olig2+/BrdU+ cells were counted by immunofluorescence. Olig1 and Olig2 gene expressions were detected by real-time fluorescent quantitative reverse transcription (qRT) -PCR. RESULTS The results showed that Catalpol improved neurological function, reduced inflammatory cell infiltration and demyelination. It could decrease Th17 cells in the peripheral blood. It increased the protein expressions of NG2 and MBP in mice brains, up-regulated markedly protein and gene expressions of Olig1 and Olig2 in terms of timing, site and targets. CONCLUSIONS These data demonstrated that Catalpol had a strong neuroprotective effect on EAE mice. Catalpol also plays a role in remyelination by promoting the expressions of Olig1 and Olig2 transcription factors.
Collapse
Affiliation(s)
- Tao Yang
- Department of Traditional Chinese Medicine, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100050, People's Republic of China
| | - Qi Zheng
- School of Traditional Chinese Medicine, Beijing Key Lab of TCM Collateral Disease Theory Research, Capital Medical University, Beijing, 100069, People's Republic of China
- Oncology Department, Guang An Men Hospital of China Academy of Chinese Medical Sciences, Beijing, 100053, People's Republic of China
| | - Su Wang
- Department of Traditional Chinese Medicine, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100050, People's Republic of China
| | - Ling Fang
- School of Traditional Chinese Medicine, Beijing Key Lab of TCM Collateral Disease Theory Research, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Lei Liu
- School of Traditional Chinese Medicine, Beijing Key Lab of TCM Collateral Disease Theory Research, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Hui Zhao
- School of Traditional Chinese Medicine, Beijing Key Lab of TCM Collateral Disease Theory Research, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Lei Wang
- School of Traditional Chinese Medicine, Beijing Key Lab of TCM Collateral Disease Theory Research, Capital Medical University, Beijing, 100069, People's Republic of China.
| | - Yongping Fan
- Department of Traditional Chinese Medicine, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100050, People's Republic of China.
| |
Collapse
|
19
|
Maduro V, Pusey BN, Cherukuri PF, Atkins P, du Souich C, Rupps R, Limbos M, Adams DR, Bhatt SS, Eydoux P, Links AE, Lehman A, Malicdan MC, Mason CE, Morimoto M, Mullikin JC, Sear A, Van Karnebeek C, Stankiewicz P, Gahl WA, Toro C, Boerkoel CF. Complex translocation disrupting TCF4 and altering TCF4 isoform expression segregates as mild autosomal dominant intellectual disability. Orphanet J Rare Dis 2016; 11:62. [PMID: 27179618 PMCID: PMC4868023 DOI: 10.1186/s13023-016-0439-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 04/25/2016] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Mutations of TCF4, which encodes a basic helix-loop-helix transcription factor, cause Pitt-Hopkins syndrome (PTHS) via multiple genetic mechanisms. TCF4 is a complex locus expressing multiple transcripts by alternative splicing and use of multiple promoters. To address the relationship between mutation of these transcripts and phenotype, we report a three-generation family segregating mild intellectual disability with a chromosomal translocation disrupting TCF4. RESULTS Using whole genome sequencing, we detected a complex unbalanced karyotype disrupting TCF4 (46,XY,del(14)(q23.3q23.3)del(18)(q21.2q21.2)del(18)(q21.2q21.2)inv(18)(q21.2q21.2)t(14;18)(q23.3;q21.2)(14pter®14q23.3::18q21.2®18q21.2::18q21.1®18qter;18pter®18q21.2::14q23.3®14qter). Subsequent transcriptome sequencing, qRT-PCR and nCounter analyses revealed that cultured skin fibroblasts and peripheral blood had normal expression of genes along chromosomes 14 or 18 and no marked changes in expression of genes other than TCF4. Affected individuals had 12-33 fold higher mRNA levels of TCF4 than did unaffected controls or individuals with PTHS. Although the derivative chromosome generated a PLEKHG3-TCF4 fusion transcript, the increased levels of TCF4 mRNA arose from transcript variants originating distal to the translocation breakpoint, not from the fusion transcript. CONCLUSIONS Although validation in additional patients is required, our findings suggest that the dysmorphic features and severe intellectual disability characteristic of PTHS are partially rescued by overexpression of those short TCF4 transcripts encoding a nuclear localization signal, a transcription activation domain, and the basic helix-loop-helix domain.
Collapse
Affiliation(s)
- Valerie Maduro
- NIH Undiagnosed Diseases Program, Common Fund, Office of the Director, National Institutes of Health, Bethesda, MD, USA
| | - Barbara N Pusey
- NIH Undiagnosed Diseases Program, Common Fund, Office of the Director, National Institutes of Health, Bethesda, MD, USA
| | - Praveen F Cherukuri
- NIH Undiagnosed Diseases Program, Common Fund, Office of the Director, National Institutes of Health, Bethesda, MD, USA
| | - Paul Atkins
- Department of Medical Genetics, University of British Columbia, Children's and Women's Health Centre of BC, Vancouver, BC, Canada
- Child and Family Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Christèle du Souich
- Department of Medical Genetics, University of British Columbia, Children's and Women's Health Centre of BC, Vancouver, BC, Canada
- Child and Family Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Rosemarie Rupps
- Department of Medical Genetics, University of British Columbia, Children's and Women's Health Centre of BC, Vancouver, BC, Canada
- Child and Family Research Institute, University of British Columbia, Vancouver, BC, Canada
| | | | - David R Adams
- NIH Undiagnosed Diseases Program, Common Fund, Office of the Director, National Institutes of Health, Bethesda, MD, USA
| | - Samarth S Bhatt
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Patrice Eydoux
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Amanda E Links
- NIH Undiagnosed Diseases Program, Common Fund, Office of the Director, National Institutes of Health, Bethesda, MD, USA
| | - Anna Lehman
- Department of Medical Genetics, University of British Columbia, Children's and Women's Health Centre of BC, Vancouver, BC, Canada
- Child and Family Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - May C Malicdan
- NIH Undiagnosed Diseases Program, Common Fund, Office of the Director, National Institutes of Health, Bethesda, MD, USA
| | - Christopher E Mason
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, NY, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, New York, NY, USA
- The Feil Family Brain and Mind Research Institute (BMRI), New York, NY, USA
| | - Marie Morimoto
- Department of Medical Genetics, University of British Columbia, Children's and Women's Health Centre of BC, Vancouver, BC, Canada
- Child and Family Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - James C Mullikin
- NIH Intramural Sequencing Center, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Andrew Sear
- Department of General Practice, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Clara Van Karnebeek
- Child and Family Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Pawel Stankiewicz
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - William A Gahl
- NIH Undiagnosed Diseases Program, Common Fund, Office of the Director, National Institutes of Health, Bethesda, MD, USA
- NHGRI, National Institutes of Health, Bethesda, MD, USA
| | - Camilo Toro
- NIH Undiagnosed Diseases Program, Common Fund, Office of the Director, National Institutes of Health, Bethesda, MD, USA
| | - Cornelius F Boerkoel
- NIH Undiagnosed Diseases Program, Common Fund, Office of the Director, National Institutes of Health, Bethesda, MD, USA.
- Department of Medical Genetics, University of British Columbia, Children's and Women's Health Centre of BC, Vancouver, BC, Canada.
- Child and Family Research Institute, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
20
|
Qi Q, Zhang Y, Shen L, Wang R, Zhou J, Lü H, Hu J. Olig1 expression pattern in neural cells during rat spinal cord development. Neuropsychiatr Dis Treat 2016; 12:909-16. [PMID: 27143892 PMCID: PMC4841409 DOI: 10.2147/ndt.s99257] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
PURPOSE Our purpose was to systematically investigate the expression pattern and role of Olig1 in neural cells during rat spinal cord development. ANIMALS AND METHODS Spinal cord tissues were dissected from Sprague-Dawley rats at embryonic day 14.5 (E14.5) and E18.5, postnatal day 0 (P0), P3, P7, postnatal 2 weeks (P2W), P4W, and adults (more than 2 months after birth), respectively. The expression of Olig1 was determined by Western blot and immunostaining. To observe expression of Olig1 in different neural cell types, a double immunohistochemical staining was performed using antibodies against Olig1 with O4, β-tubulin, glial fibrillary acidic protein (GFAP), and myelin basic protein, respectively. RESULTS The expression of Olig1 protein shows a significant level change in rat spinal cord at different developmental time points. Starting with E14.5, the expression gradually increased and peaked at E18.5. Olig1 decreased gradually from P3 and reached its lowest level on P7. However, interestingly, the Olig1 expression increased again from P2W, until adulthood. Olig1 was coexpressed with O4-positive oligodendrocyte progenitor cells (OPCs) and β-tubulin-positive neurons at all time points during development. Olig1 was also coexpressed transiently with GFAP-positive astrocytes at only E14.5. Olig1 was localized in the cytoplasm of O4- and β-tubulin-positive cells during the period from E14.5 to adult. CONCLUSION The expression of Olig1 in OPCs and neurons at all time points during development and in astrocytes at E14.5 suggests that Olig1 may play an important role in the generation and maturation of specific neural cells during development of spinal cord. Our results contribute to understanding the mechanism underlying developmental regulation of neural cells by Olig1.
Collapse
Affiliation(s)
- Qi Qi
- Anhui Key Laboratory of Tissue Transplantation, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, People's Republic of China; Department of Histology and Embryology, Bengbu Medical College, Bengbu, Anhui, People's Republic of China
| | - Yuxin Zhang
- Anhui Key Laboratory of Tissue Transplantation, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, People's Republic of China
| | - Lin Shen
- Anhui Key Laboratory of Tissue Transplantation, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, People's Republic of China
| | - Rui Wang
- Anhui Key Laboratory of Tissue Transplantation, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, People's Republic of China
| | - Jiansheng Zhou
- Anhui Key Laboratory of Tissue Transplantation, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, People's Republic of China
| | - Hezuo Lü
- Anhui Key Laboratory of Tissue Transplantation, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, People's Republic of China; Department of Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, People's Republic of China
| | - Jianguo Hu
- Anhui Key Laboratory of Tissue Transplantation, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, People's Republic of China; Department of Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, People's Republic of China
| |
Collapse
|
21
|
Wang W, Liu R, Xu Z, Niu X, Mao Z, Meng Q, Cao X. Further insight into molecular mechanism underlying thoracic spinal cord injury using bioinformatics methods. Mol Med Rep 2015; 12:7851-8. [PMID: 26497545 PMCID: PMC4758289 DOI: 10.3892/mmr.2015.4442] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 04/24/2015] [Indexed: 02/02/2023] Open
Abstract
The present study aimed to explore the molecular mechanisms underlying the development of thoracic spinal cord injury (SCI). The gene expression profile of GSE20907, which included 12 thoracic non-injured spinal cord control samples and 12 thoracic transected spinal cord samples at different stages of SCI, was obtained from the Gene Expression Omnibus database. Differentially expressed genes (DEGs) were identified using the limma package in R/Bioconductor. DEG-associated pathways were analyzed using the Kyoto encyclopedia of genes and genomes database. A protein-protein interaction (PPI) network was constructed and transcription factors (TFs) were predicted using cytoscape. Compared with the control samples, there were 1,942, 396, 188 and 396 DEGs identified at day 3 (d3), week 1 (wk1), wk2 and month 1 (m1), respectively. Cluster analysis indicated that the DEGs at m1 were similar to those in the control group. Downregulated DEGs were enriched in nervous system disease pathways, such as Parkinson's disease. Upregulated DEGs were enriched in immune response-associated pathways, such as Fc γ R-mediated phagocytosis at early stages (d3 and wk1). Upregulated DEGs were enriched in pathways associated with cancer and pyrimidine metabolism at wk2 and m1, respectively. In the PPI network, nodes including RAC2, CD4, STAT3 and JUN were identified. Furthermore, ATF3, JUN and EGR1 were identified as TFs associated with SCI. In conclusion, the results of the present study showed that the number of DEGs decreased in a time-dependent manner following SCI. OLIG1, ATF3 and JUN may represent SCI regeneration-associated genes. Immune-associated inflammation was shown to be important in SCI, and SCI exhibits causal associations with other diseases, including cardiovascular disease and cancers. The present study provided novel insight into the molecular mechanisms of SCI regeneration, which may aid in the development of strategies to enhance recovery following SCI.
Collapse
Affiliation(s)
- Weiguo Wang
- Department of Orthopaedic Surgery, General Hospital of Jinan Military Command, Jinan, Shandong 250031, P.R. China
| | - Rongjun Liu
- Department of Emergency Surgery, General Hospital of Jinan Military Command, Jinan, Shandong 250031, P.R. China
| | - Zhanwang Xu
- Department of Orthopedics, First Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250014, P.R. China
| | - Xiufeng Niu
- Department of Hepatobiliary Surgery, General Hospital of Jinan Military Command, Jinan, Shandong 250031, P.R. China
| | - Zhaohu Mao
- Department of Spinal Cord Injury, General Hospital of Jinan Military Command, Jinan, Shandong 250031, P.R. China
| | - Qingxi Meng
- Department of Spinal Cord Injury, General Hospital of Jinan Military Command, Jinan, Shandong 250031, P.R. China
| | - Xuecheng Cao
- Department of Orthopaedic Surgery, General Hospital of Jinan Military Command, Jinan, Shandong 250031, P.R. China
| |
Collapse
|
22
|
Praet J, Guglielmetti C, Berneman Z, Van der Linden A, Ponsaerts P. Cellular and molecular neuropathology of the cuprizone mouse model: clinical relevance for multiple sclerosis. Neurosci Biobehav Rev 2015; 47:485-505. [PMID: 25445182 DOI: 10.1016/j.neubiorev.2014.10.004] [Citation(s) in RCA: 281] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 09/18/2014] [Accepted: 10/01/2014] [Indexed: 01/30/2023]
Abstract
The cuprizone mouse model allows the investigation of the complex molecular mechanisms behind nonautoimmune-mediated demyelination and spontaneous remyelination. While it is generally accepted that oligodendrocytes are specifically vulnerable to cuprizone intoxication due to their high metabolic demands, a comprehensive overview of the etiology of cuprizone-induced pathology is still missing to date. In this review we extensively describe the physico-chemical mode of action of cuprizone and discuss the molecular and enzymatic mechanisms by which cuprizone induces metabolic stress, oligodendrocyte apoptosis, myelin degeneration and eventually axonal and neuronal pathology. In addition, we describe the dual effector function of the immune system which tightly controls demyelination by effective induction of oligodendrocyte apoptosis, but in contrast also paves the way for fast and efficient remyelination by the secretion of neurotrophic factors and the clearance of cellular and myelinic debris. Finally, we discuss the many clinical symptoms that can be observed following cuprizone treatment, and how these strengthened the cuprizone model as a useful tool to study human multiple sclerosis, schizophrenia and epilepsy.
Collapse
|
23
|
Intracellular Protein Shuttling: A Mechanism Relevant for Myelin Repair in Multiple Sclerosis? Int J Mol Sci 2015; 16:15057-85. [PMID: 26151843 PMCID: PMC4519887 DOI: 10.3390/ijms160715057] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 06/23/2015] [Accepted: 06/25/2015] [Indexed: 12/15/2022] Open
Abstract
A prominent feature of demyelinating diseases such as multiple sclerosis (MS) is the degeneration and loss of previously established functional myelin sheaths, which results in impaired signal propagation and axonal damage. However, at least in early disease stages, partial replacement of lost oligodendrocytes and thus remyelination occur as a result of resident oligodendroglial precursor cell (OPC) activation. These cells represent a widespread cell population within the adult central nervous system (CNS) that can differentiate into functional myelinating glial cells to restore axonal functions. Nevertheless, the spontaneous remyelination capacity in the adult CNS is inefficient because OPCs often fail to generate new oligodendrocytes due to the lack of stimulatory cues and the presence of inhibitory factors. Recent studies have provided evidence that regulated intracellular protein shuttling is functionally involved in oligodendroglial differentiation and remyelination activities. In this review we shed light on the role of the subcellular localization of differentiation-associated factors within oligodendroglial cells and show that regulation of intracellular localization of regulatory factors represents a crucial process to modulate oligodendroglial maturation and myelin repair in the CNS.
Collapse
|
24
|
Yang W, Xiao L, Li C, Liu X, Liu M, Shao Q, Wang D, Huang A, He C. TIP30 inhibits oligodendrocyte precursor cell differentiation via cytoplasmic sequestration of Olig1. Glia 2014; 63:684-98. [PMID: 25530119 DOI: 10.1002/glia.22778] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2014] [Revised: 11/27/2014] [Accepted: 12/03/2014] [Indexed: 12/24/2022]
Abstract
Differentiation of oligodendrocyte precursor cells (OPCs) is a prerequisite for both developmental myelination and adult remyelination in the central nervous system. The molecular mechanisms underlying OPC differentiation remain largely unknown. Here, we show that the thirty-kDa HIV-1 Tat interacting protein (TIP30) is a negative regulator in oligodendrocyte development. The TIP30(-/-) mice displayed an increased myelin protein level at postnatal day 14 and 21. By using a primary OPC culture system, we demonstrated that overexpression of TIP30 dramatically inhibited the stage progression of differentiating OPCs, while knockdown of TIP30 enhanced the differentiation of oligodendroglial cells remarkably. Moreover, overexpression of TIP30 was found to sequester the transcription factor Olig1 in the cytoplasm and weaken its nuclear translocation due to the interaction between TIP30 and Olig1, whereas knockdown of TIP30 led to more Olig1 localized in the nucleus in the initiation stage during OPC differentiation. In the cuprizone-induced demyelination model, there was a dramatic increase in NG2-expressing cells with nuclear location of Olig1 in the corpus callosum during remyelination. In contrast, within chronic demyelinated lesions in multiple sclerosis, TIP30 was abnormally expressed in NG2-expressing cells, and few nuclear Olig1 was observed in these cells. Taken together, our findings suggest that TIP30 plays a negative regulatory role in oligodendroglial differentiation.
Collapse
Affiliation(s)
- Wenjing Yang
- Institute of Neuroscience and MOE Key Laboratory of Molecular Neurobiology, Neuroscience Research Center of Changzheng Hospital, Second Military Medical University, Shanghai, China
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Czepiel M, Boddeke E, Copray S. Human oligodendrocytes in remyelination research. Glia 2014; 63:513-30. [DOI: 10.1002/glia.22769] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 11/05/2014] [Indexed: 01/04/2023]
Affiliation(s)
- Marcin Czepiel
- Department of Neuroscience; University Medical Center Groningen; A.Deusinglaan 1, 9713AV Groningen The Netherlands
| | - Erik Boddeke
- Department of Neuroscience; University Medical Center Groningen; A.Deusinglaan 1, 9713AV Groningen The Netherlands
| | - Sjef Copray
- Department of Neuroscience; University Medical Center Groningen; A.Deusinglaan 1, 9713AV Groningen The Netherlands
| |
Collapse
|
26
|
Cui H, Han W, Yang L, Chang Y. Expression of hypoxia-inducible factor 1 alpha and oligodendrocyte lineage gene-1 in cultured brain slices after oxygen-glucose deprivation. Neural Regen Res 2014; 8:328-37. [PMID: 25206673 PMCID: PMC4107529 DOI: 10.3969/j.issn.1673-5374.2013.04.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2012] [Accepted: 01/09/2013] [Indexed: 02/03/2023] Open
Abstract
Oligodendrocyte lineage gene-1 expressed in oligodendrocytes may trigger the repair of neuronal myelin impairment, and play a crucial role in myelin repair. Hypoxia-inducible factor 1α, a transcription factor, is of great significance in premature infants with hypoxic-ischemic brain damage. There is little evidence of direct regulatory effects of hypoxia-inducible factor 1α on oligodendrocyte lineage gene-1. In this study, brain slices of Sprague-Dawley rats were cultured and subjected to oxygen-glucose deprivation. Then, slices were transfected with hypoxia-inducible factor 1α or oligodendrocyte lineage gene-1. The expression levels of hypoxia-inducible factor 1α and oligodendrocyte lineage gene-1 were significantly up-regulated in rat brains prior to transfection, as detected by immunohistochemical staining. Eight hours after transfection of slices with hypoxia-inducible factor 1α, oligodendrocyte lineage gene-1 expression was upregulated, and reached a peak 24 hours after transfection. Oligodendrocyte lineage gene-1 transfection induced no significant differences in hypoxia-inducible factor 1α levels in rat brain tissues with oxygen-glucose deprivation. These experimental findings indicate that hypoxia-inducible factor 1α can regulate oligodendrocyte lineage gene-1 expression in hypoxic brain tissue, thus repairing the neural impairment.
Collapse
Affiliation(s)
- Hong Cui
- College of Life Science, Hebei Normal University, Shijiazhuang 050016, Hebei Province, China ; Department of Pediatrics, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Weijuan Han
- Department of Pediatrics, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Lijun Yang
- Department of Pediatrics, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Yanzhong Chang
- College of Life Science, Hebei Normal University, Shijiazhuang 050016, Hebei Province, China
| |
Collapse
|
27
|
Tan B, Yu J, Yin Y, Jia G, Jiang W, Yu L. The Olig family affects central nervous system development and disease. Neural Regen Res 2014; 9:329-36. [PMID: 25206819 PMCID: PMC4146145 DOI: 10.4103/1673-5374.128232] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/15/2013] [Indexed: 11/04/2022] Open
Abstract
Neural cell differentiation and maturation is a critical step during central nervous system development. The oligodendrocyte transcription family (Olig family) is known to be an important factor in regulating neural cell differentiation. Because of this, the Olig family also affects acute and chronic central nervous system diseases, including brain injury, multiple sclerosis, and even gliomas. Improved understanding about the functions of the Olig family in central nervous system development and disease will greatly aid novel breakthroughs in central nervous system diseases. This review investigates the role of the Olig family in central nervous system development and related diseases.
Collapse
Affiliation(s)
- Botao Tan
- Department of Rehabilitation Medicine, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Jing Yu
- Department of Rehabilitation Medicine, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Ying Yin
- Department of Rehabilitation Medicine, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Gongwei Jia
- Department of Rehabilitation Medicine, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Wei Jiang
- Department of Rehabilitation Medicine, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Lehua Yu
- Department of Rehabilitation Medicine, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| |
Collapse
|
28
|
Baxi EG, Schott JT, Fairchild AN, Kirby LA, Karani R, Uapinyoying P, Pardo-Villamizar C, Rothstein JR, Bergles DE, Calabresi PA. A selective thyroid hormone β receptor agonist enhances human and rodent oligodendrocyte differentiation. Glia 2014; 62:1513-29. [PMID: 24863526 DOI: 10.1002/glia.22697] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Revised: 04/25/2014] [Accepted: 05/07/2014] [Indexed: 01/16/2023]
Abstract
Nerve conduction within the mammalian central nervous system is made efficient by oligodendrocyte-derived myelin. Historically, thyroid hormones have a well described role in regulating oligodendrocyte differentiation and myelination during development; however, it remains unclear which thyroid hormone receptors are required to drive these effects. This is a question with clinical relevance since nonspecific thyroid receptor stimulation can produce deleterious side-effects. Here we report that GC-1, a thyromimetic with selective thyroid receptor β action and a potentially limited side-effect profile, promotes in vitro oligodendrogenesis from both rodent and human oligodendrocyte progenitor cells. In addition, we used in vivo genetic fate tracing of oligodendrocyte progenitor cells via PDGFαR-CreER;Rosa26-eYFP double-transgenic mice to examine the effect of GC-1 on cellular fate and find that treatment with GC-1 during developmental myelination promotes oligodendrogenesis within the corpus callosum, occipital cortex and optic nerve. GC-1 was also observed to enhance the expression of the myelin proteins MBP, CNP and MAG within the same regions. These results indicate that a β receptor selective thyromimetic can enhance oligodendrocyte differentiation in vitro and during developmental myelination in vivo and warrants further study as a therapeutic agent for demyelinating models.
Collapse
Affiliation(s)
- Emily G Baxi
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Schmid D, Zeis T, Schaeren-Wiemers N. Transcriptional regulation induced by cAMP elevation in mouse Schwann cells. ASN Neuro 2014; 6:137-57. [PMID: 24641305 PMCID: PMC4834722 DOI: 10.1042/an20130031] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Revised: 01/16/2014] [Accepted: 02/05/2014] [Indexed: 12/23/2022] Open
Abstract
In peripheral nerves, Schwann cell development is regulated by a variety of signals. Some of the aspects of Schwann cell differentiation can be reproduced in vitro in response to forskolin, an adenylyl cyclase activator elevating intracellular cAMP levels. Herein, the effect of forskolin treatment was investigated by a comprehensive genome-wide expression study on primary mouse Schwann cell cultures. Additional to myelin-related genes, many so far unconsidered genes were ascertained to be modulated by forskolin. One of the strongest differentially regulated gene transcripts was the transcription factor Olig1 (oligodendrocyte transcription factor 1), whose mRNA expression levels were reduced in treated Schwann cells. Olig1 protein was localized in myelinating and nonmyelinating Schwann cells within the sciatic nerve as well as in primary Schwann cells, proposing it as a novel transcription factor of the Schwann cell lineage. Data analysis further revealed that a number of differentially expressed genes in forskolin-treated Schwann cells were associated with the ECM (extracellular matrix), underlining its importance during Schwann cell differentiation in vitro. Comparison of samples derived from postnatal sciatic nerves and from both treated and untreated Schwann cell cultures showed considerable differences in gene expression between in vivo and in vitro, allowing us to separate Schwann cell autonomous from tissue-related changes. The whole data set of the cell culture microarray study is provided to offer an interactive search tool for genes of interest.
Collapse
Key Words
- camp
- forskolin
- in vitro
- microarray
- schwann cell differentiation
- bmp, bone morphogenetic protein
- camp, cyclic adenosine monophosphate
- cns, central nervous system
- creb, camp-response-element-binding protein
- david, database for annotation, visualization and integrated discovery
- dgc, dystrophin–glycoprotein complex
- ecm, extracellular matrix
- fdr, false discovery rate
- go, gene ontology
- ipa, ingenuity pathway analysis
- mag, myelin-associated glycoprotein
- mapk, mitogen-activated protein kinase
- mbp, myelin basic protein
- mpz/p0, myelin protein zero
- nf-κb, nuclear factor κb
- olig1, oligodendrocyte transcription factor 1
- pca, principal component analysis
- pfa, paraformaldehyde
- pka, protein kinase a
- pns, peripheral nervous system
- qrt–pcr, quantitative rt–pcr
- s.d., standard deviation
Collapse
Affiliation(s)
- Daniela Schmid
- *Neurobiology, Department of Biomedicine, University Hospital Basel,
University of Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland
| | - Thomas Zeis
- *Neurobiology, Department of Biomedicine, University Hospital Basel,
University of Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland
| | - Nicole Schaeren-Wiemers
- *Neurobiology, Department of Biomedicine, University Hospital Basel,
University of Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland
| |
Collapse
|
30
|
Kucharska-Mazur J, Tarnowski M, Dołęgowska B, Budkowska M, Pędziwiatr D, Jabłoński M, Pełka-Wysiecka J, Kazimierczak A, Ratajczak MZ, Samochowiec J. Novel evidence for enhanced stem cell trafficking in antipsychotic-naïve subjects during their first psychotic episode. J Psychiatr Res 2014; 49:18-24. [PMID: 24246416 DOI: 10.1016/j.jpsychires.2013.10.016] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2013] [Revised: 10/23/2013] [Accepted: 10/23/2013] [Indexed: 12/18/2022]
Abstract
In this study, we tested the novel hypothesis that stem cells and those factors that modulate their trafficking may be biological markers for acute psychosis. Twenty-eight subjects during their first nonaffective psychotic episode were investigated before and after antipsychotic treatment and were compared with 35 healthy controls (CG); the psychotic group (PG) was divided into "schizophrenic" (SG) and "non-schizophrenic" (NG) subgroups. We examined the number of circulating Lin(-)/CD45(-)/CD34(+) and Lin(-)/CD45(-)/CD133(+) very small embryonic-like stem cells (VSELs), which express markers of the neural lineage, and also the plasma levels of factors that modulate their trafficking: the C3a, C5a, and C5b-9 activated complement cascade components, stromal-derived factor 1, and sphingosine-1-phosphate (S1P). We found that the mean numbers of Lin(-)/CD45(-)/CD34(+) VSELs and the plasma levels of S1P prior to treatment differ between the CG and PG and that these cells express markers of neural lineage. The number of Lin(-)/CD45(-)/CD133(+) VSELs in peripheral blood differed between the SG and NG prior to treatment. Using logistic regression analysis, we found that C3a and S1P are the best predictors of risk and are potential markers for the first psychotic episode. Furthermore, in the SG, the number of circulating Lin(-)/CD45(-)/CD34(+) VSELs and the S1P plasma level are the best predictors of risk and are proposed as novel markers for the first "schizophrenic" episode of psychosis.
Collapse
Affiliation(s)
| | - Maciej Tarnowski
- Department of Physiology, Pomeranian University of Medicine, Szczecin, Poland
| | - Barbara Dołęgowska
- Department of Medical Analytics, Pomeranian University of Medicine, Szczecin, Poland
| | - Marta Budkowska
- Department of Medical Analytics, Pomeranian University of Medicine, Szczecin, Poland
| | - Daniel Pędziwiatr
- Department of Physiology, Pomeranian University of Medicine, Szczecin, Poland
| | - Marcin Jabłoński
- Department of Psychiatry, Pomeranian University of Medicine, Szczecin, Poland
| | | | | | - Mariusz Z Ratajczak
- Department of Physiology, Pomeranian University of Medicine, Szczecin, Poland; Stem Cell Biology Program at the James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA
| | - Jerzy Samochowiec
- Department of Psychiatry, Pomeranian University of Medicine, Szczecin, Poland.
| |
Collapse
|
31
|
Kaiser O, Aliuos P, Wissel K, Lenarz T, Werner D, Reuter G, Kral A, Warnecke A. Dissociated neurons and glial cells derived from rat inferior colliculi after digestion with papain. PLoS One 2013; 8:e80490. [PMID: 24349001 PMCID: PMC3861243 DOI: 10.1371/journal.pone.0080490] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Accepted: 10/13/2013] [Indexed: 01/10/2023] Open
Abstract
The formation of gliosis around implant electrodes for deep brain stimulation impairs electrode–tissue interaction. Unspecific growth of glial tissue around the electrodes can be hindered by altering physicochemical material properties. However, in vitro screening of neural tissue–material interaction requires an adequate cell culture system. No adequate model for cells dissociated from the inferior colliculus (IC) has been described and was thus the aim of this study. Therefore, IC were isolated from neonatal rats (P3_5) and a dissociated cell culture was established. In screening experiments using four dissociation methods (Neural Tissue Dissociation Kit [NTDK] T, NTDK P; NTDK PN, and a validated protocol for the dissociation of spiral ganglion neurons [SGN]), the optimal media, and seeding densities were identified. Thereafter, a dissociation protocol containing only the proteolytic enzymes of interest (trypsin or papain) was tested. For analysis, cells were fixed and immunolabeled using glial- and neuron-specific antibodies. Adhesion and survival of dissociated neurons and glial cells isolated from the IC were demonstrated in all experimental settings. Hence, preservation of type-specific cytoarchitecture with sufficient neuronal networks only occurred in cultures dissociated with NTDK P, NTDK PN, and fresh prepared papain solution. However, cultures obtained after dissociation with papain, seeded at a density of 2×104 cells/well and cultivated with Neuro Medium for 6 days reliably revealed the highest neuronal yield with excellent cytoarchitecture of neurons and glial cells. The herein described dissociated culture can be utilized as in vitro model to screen interactions between cells of the IC and surface modifications of the electrode.
Collapse
Affiliation(s)
- Odett Kaiser
- Department of Otolaryngology, Hannover Medical School, Hannover, Germany
| | - Pooyan Aliuos
- Department of Otolaryngology, Hannover Medical School, Hannover, Germany
| | - Kirsten Wissel
- Department of Otolaryngology, Hannover Medical School, Hannover, Germany
| | - Thomas Lenarz
- Department of Otolaryngology, Hannover Medical School, Hannover, Germany
| | - Darja Werner
- Department of Otolaryngology, Hannover Medical School, Hannover, Germany
| | - Günter Reuter
- Department of Otolaryngology, Hannover Medical School, Hannover, Germany
| | - Andrej Kral
- Department of Otolaryngology, Hannover Medical School, Hannover, Germany
| | - Athanasia Warnecke
- Department of Otolaryngology, Hannover Medical School, Hannover, Germany
- * E-mail:
| |
Collapse
|
32
|
Medina-Rodríguez EM, Arenzana FJ, Bribián A, de Castro F. Protocol to isolate a large amount of functional oligodendrocyte precursor cells from the cerebral cortex of adult mice and humans. PLoS One 2013; 8:e81620. [PMID: 24303061 PMCID: PMC3841116 DOI: 10.1371/journal.pone.0081620] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Accepted: 10/24/2013] [Indexed: 01/09/2023] Open
Abstract
During development, oligodendrocytes are generated from oligodendrocyte precursor cells (OPCs), a cell type that is a significant proportion of the total cells (3-8%) in the adult central nervous system (CNS) of both rodents and humans. Adult OPCs are responsible for the spontaneous remyelination that occurs in demyelinating diseases like Multiple Sclerosis (MS) and they constitute an interesting source of cells for regenerative therapy in such conditions. However, there is little data regarding the neurobiology of adult OPCs isolated from mice since an efficient method to isolate them has yet to be established. We have designed a protocol to obtain viable adult OPCs from the cerebral cortex of different mouse strains and we have compared its efficiency with other well-known methods. In addition, we show that this protocol is also useful to isolate functional OPCs from human brain biopsies. Using this method we can isolate primary cortical OPCs in sufficient quantities so as to be able to study their survival, maturation and function, and to facilitate an evaluation of their utility in myelin repair.
Collapse
Affiliation(s)
| | | | - Ana Bribián
- Grupo de Neurobiología del Desarrollo-GNDe, Hospital Nacional de Parapléjicos, Toledo, Spain
| | - Fernando de Castro
- Grupo de Neurobiología del Desarrollo-GNDe, Hospital Nacional de Parapléjicos, Toledo, Spain
- * E-mail:
| |
Collapse
|
33
|
Xu H, Belkacemi L, Jog M, Parrent A, Hebb MO. Neurotrophic factor expression in expandable cell populations from brain samples in living patients with Parkinson's disease. FASEB J 2013; 27:4157-68. [PMID: 23825231 DOI: 10.1096/fj.12-226555] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Cell-based therapies offer promise for patients with Parkinson's disease (PD); however, durable and effective transplantation substrates need to be defined. This study characterized the feasibility and growth properties of primary cultures established from small-volume brain biopsies taken during deep brain stimulation (DBS) surgery in patients with PD. The lineage and expression of neurotrophic factors with known beneficial actions in PD-affected brain circuitry were also evaluated. Nineteen patients with PD undergoing DBS surgery consented to brain biopsies prior to electrode implantation. Cultures from these samples exhibited exponential and plateau phases of growth and were readily expanded throughout multiple passages. There was robust expression of progenitor markers and the unexpected colocalization of neural and mesenchymal proteins. The oligodendrocyte transcription factor, Olig1, and the myelin-specific sphingolipid, galactocerebroside, were coexpressed with each of glial-derived neurotrophic factor, brain-derived neurotrophic factor, and cerebral dopamine neurotrophic factor. Fluorescence-activated cell sorting demonstrated homogeneous expression of both nestin and Olig1 throughout the expanded cultures. Cells remained viable after a year in cryostorage. These findings confirm the feasibility of small brain biopsies as an expandable source of autologous cell substrate in living patients and demonstrate the complex phenotype of these cells, with implications for therapeutic application in PD and other neurological diseases.
Collapse
Affiliation(s)
- Hu Xu
- 1Department of Clinical Neurological Sciences (Neurosurgery), University of Western Ontario, 339 Windermere Rd., C7-134, London, ON, Canada N6A 5A5.
| | | | | | | | | |
Collapse
|
34
|
Szymanski CR, Chiha W, Morellini N, Cummins N, Bartlett CA, O'Hare Doig RL, Savigni DL, Payne SC, Harvey AR, Dunlop SA, Fitzgerald M. Paranode Abnormalities and Oxidative Stress in Optic Nerve Vulnerable to Secondary Degeneration: Modulation by 670 nm Light Treatment. PLoS One 2013; 8:e66448. [PMID: 23840470 PMCID: PMC3686728 DOI: 10.1371/journal.pone.0066448] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Accepted: 05/05/2013] [Indexed: 11/25/2022] Open
Abstract
Secondary degeneration of nerve tissue adjacent to a traumatic injury results in further loss of neurons, glia and function, via mechanisms that may involve oxidative stress. However, changes in indicators of oxidative stress have not yet been demonstrated in oligodendrocytes vulnerable to secondary degeneration in vivo. We show increases in the oxidative stress indicator carboxymethyl lysine at days 1 and 3 after injury in oligodendrocytes vulnerable to secondary degeneration. Dihydroethidium staining for superoxide is reduced, indicating endogenous control of this particular reactive species after injury. Concurrently, node of Ranvier/paranode complexes are altered, with significant lengthening of the paranodal gap and paranode as well as paranode disorganisation. Therapeutic administration of 670 nm light is thought to improve oxidative metabolism via mechanisms that may include increased activity of cytochrome c oxidase. Here, we show that light at 670 nm, delivered for 30 minutes per day, results in in vivo increases in cytochrome c oxidase activity co-localised with oligodendrocytes. Short term (1 day) 670 nm light treatment is associated with reductions in reactive species at the injury site. In optic nerve vulnerable to secondary degeneration superoxide in oligodendrocytes is reduced relative to handling controls, and is associated with reduced paranode abnormalities. Long term (3 month) administration of 670 nm light preserves retinal ganglion cells vulnerable to secondary degeneration and maintains visual function, as assessed by the optokinetic nystagmus visual reflex. Light at a wavelength of 670 nm may serve as a therapeutic intervention for treatment of secondary degeneration following neurotrauma.
Collapse
Affiliation(s)
- Charis R. Szymanski
- Experimental and Regenerative Neurosciences, School of Animal Biology, The University of Western Australia, Crawley, Western Australia, Australia
| | - Wissam Chiha
- Experimental and Regenerative Neurosciences, School of Animal Biology, The University of Western Australia, Crawley, Western Australia, Australia
| | - Natalie Morellini
- Experimental and Regenerative Neurosciences, School of Animal Biology, The University of Western Australia, Crawley, Western Australia, Australia
| | - Nadia Cummins
- Experimental and Regenerative Neurosciences, School of Animal Biology, The University of Western Australia, Crawley, Western Australia, Australia
| | - Carole A. Bartlett
- Experimental and Regenerative Neurosciences, School of Animal Biology, The University of Western Australia, Crawley, Western Australia, Australia
| | - Ryan L. O'Hare Doig
- Experimental and Regenerative Neurosciences, School of Animal Biology, The University of Western Australia, Crawley, Western Australia, Australia
| | - Donna L. Savigni
- Experimental and Regenerative Neurosciences, School of Animal Biology, The University of Western Australia, Crawley, Western Australia, Australia
| | - Sophie C. Payne
- Experimental and Regenerative Neurosciences, School of Animal Biology, The University of Western Australia, Crawley, Western Australia, Australia
| | - Alan R. Harvey
- Experimental and Regenerative Neurosciences, School of Anatomy, Physiology and Human Biology, The University of Western Australia, Crawley, Western Australia, Australia
| | - Sarah A. Dunlop
- Experimental and Regenerative Neurosciences, School of Animal Biology, The University of Western Australia, Crawley, Western Australia, Australia
| | - Melinda Fitzgerald
- Experimental and Regenerative Neurosciences, School of Animal Biology, The University of Western Australia, Crawley, Western Australia, Australia
- * E-mail:
| |
Collapse
|
35
|
Navarrete K, Pedroso I, De Jong S, Stefansson H, Steinberg S, Stefansson K, Ophoff RA, Schalkwyk LC, Collier DA. TCF4 (e2-2; ITF2): a schizophrenia-associated gene with pleiotropic effects on human disease. Am J Med Genet B Neuropsychiatr Genet 2013; 162B:1-16. [PMID: 23129290 DOI: 10.1002/ajmg.b.32109] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2012] [Accepted: 09/27/2012] [Indexed: 12/22/2022]
Abstract
Common SNPs in the transcription factor 4 (TCF4; ITF2, E2-2, SEF-2) gene, which encodes a basic Helix-Loop-Helix (bHLH) transcription factor, are associated with schizophrenia, conferring a small increase in risk. Other common SNPs in the gene are associated with the common eye disorder Fuch's corneal dystrophy, while rare, mostly de novo inactivating mutations cause Pitt-Hopkins syndrome. In this review, we present a systematic bioinformatics and literature review of the genomics, biological function and interactome of TCF4 in the context of schizophrenia. The TCF4 gene is present in all vertebrates, and although protein length varies, there is high conservation of primary sequence, including the DNA binding domain. Humans have a unique leucine-rich nuclear export signal. There are two main isoforms (A and B), as well as complex splicing generating many possible N-terminal amino acid sequences. TCF4 is highly expressed in the brain, where plays a role in neurodevelopment, interacting with class II bHLH transcription factors Math1, HASH1, and neuroD2. The Ca(2+) sensor protein calmodulin interacts with the DNA binding domain of TCF4, inhibiting transcriptional activation. It is also the target of microRNAs, including mir137, which is implicated in schizophrenia. The schizophrenia-associated SNPs are in linkage disequilibrium with common variants within putative DNA regulatory elements, suggesting that regulation of expression may underlie association with schizophrenia. Combined gene co-expression analyses and curated protein-protein interaction data provide a network involving TCF4 and other putative schizophrenia susceptibility genes. These findings suggest new opportunities for understanding the molecular basis of schizophrenia and other mental disorders.
Collapse
Affiliation(s)
- Katherinne Navarrete
- Social, Genetic and Developmental Psychiatry Centre, King's College London, Institute of Psychiatry, London, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Barateiro A, Miron VE, Santos SD, Relvas JB, Fernandes A, ffrench-Constant C, Brites D. Unconjugated Bilirubin Restricts Oligodendrocyte Differentiation and Axonal Myelination. Mol Neurobiol 2012; 47:632-44. [DOI: 10.1007/s12035-012-8364-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Accepted: 10/08/2012] [Indexed: 01/05/2023]
|
37
|
Niu J, Mei F, Wang L, Liu S, Tian Y, Mo W, Li H, Lu QR, Xiao L. Phosphorylated olig1 localizes to the cytosol of oligodendrocytes and promotes membrane expansion and maturation. Glia 2012; 60:1427-36. [PMID: 22639060 DOI: 10.1002/glia.22364] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2012] [Revised: 04/03/2012] [Accepted: 05/07/2012] [Indexed: 11/11/2022]
Abstract
Oligodendroglial cells undergo rapid transcriptional and dynamic morphological transformation in order to effectively myelinate neuronal axons. Olig1, a basic helix-loop-helix transcription factor, functions to promote the transcription of myelin-specific genes and promotes differentiation and (re)myelination. While the role for nuclear Olig1 is well established, the function for cytoplasmic Olig1 remains uncertain. We observe that translocation of Olig1 into the cytosol highly correlates with differentiation of oligodendrocytes both invivo and invitro. By enforcing expression of a nuclear-specific form of Olig1 into OPCs in a null-Olig1 background, we demonstrate that nuclear Olig1 is sufficient to facilitate MBP expression, but with greatly diminished membrane volume and area. We demonstrate that serine 138 in the helix-loop-helix domain of Olig1 is phosphorylated and that this form resides in the cytosol. Mutating serine 138 to alanine restricts Olig1 to the nucleus, facilitating MBP expression but limiting membrane expansion. However, a serine to aspartic acid mutation results in the cytoplasmic localization of Olig1 enhancing membrane expansion. Our results suggest a novel role for a phosphorylated cytosolic Olig1 in membrane expansion and maturation of oligodendrocytes.
Collapse
Affiliation(s)
- Jianqin Niu
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Third Military Medical University, Chongqing, China
| | | | | | | | | | | | | | | | | |
Collapse
|