1
|
Ma Z, Zhang W, Wang C, Su Y, Yi C, Niu J. A New Acquaintance of Oligodendrocyte Precursor Cells in the Central Nervous System. Neurosci Bull 2024; 40:1573-1589. [PMID: 39042298 PMCID: PMC11422404 DOI: 10.1007/s12264-024-01261-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 03/21/2024] [Indexed: 07/24/2024] Open
Abstract
Oligodendrocyte precursor cells (OPCs) are a heterogeneous multipotent population in the central nervous system (CNS) that appear during embryogenesis and persist as resident cells in the adult brain parenchyma. OPCs could generate oligodendrocytes to participate in myelination. Recent advances have renewed our knowledge of OPC biology by discovering novel markers of oligodendroglial cells, the myelin-independent roles of OPCs, and the regulatory mechanism of OPC development. In this review, we will explore the updated knowledge on OPC identity, their multifaceted roles in the CNS in health and diseases, as well as the regulatory mechanisms that are involved in their developmental stages, which hopefully would contribute to a further understanding of OPCs and attract attention in the field of OPC biology.
Collapse
Affiliation(s)
- Zexuan Ma
- Department of Histology and Embryology, College of basic medicine, Third Military Medical University, Chongqing, 400038, China
| | - Wei Zhang
- Department of Histology and Embryology, College of basic medicine, Third Military Medical University, Chongqing, 400038, China
| | - Chenmeng Wang
- Department of Histology and Embryology, College of basic medicine, Third Military Medical University, Chongqing, 400038, China
- Research Centre, Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Yixun Su
- Research Centre, Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Chenju Yi
- Research Centre, Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China.
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangzhou, China.
- Shenzhen Key Laboratory of Chinese Medicine Active substance screening and Translational Research, Shenzhen, 518107, China.
| | - Jianqin Niu
- Department of Histology and Embryology, College of basic medicine, Third Military Medical University, Chongqing, 400038, China.
- Chongqing Key Laboratory of Neurobiology, Chongqing, 400038, China.
| |
Collapse
|
2
|
Jia R, Ren YZ, Li PN, Gao R, Zhang YS. SCSMD: Single Cell Consistent Clustering based on Spectral Matrix Decomposition. Brief Bioinform 2024; 25:bbae273. [PMID: 38855914 PMCID: PMC11163303 DOI: 10.1093/bib/bbae273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 04/25/2024] [Accepted: 05/30/2024] [Indexed: 06/11/2024] Open
Abstract
Cluster analysis, a pivotal step in single-cell sequencing data analysis, presents substantial opportunities to effectively unveil the molecular mechanisms underlying cellular heterogeneity and intercellular phenotypic variations. However, the inherent imperfections arise as different clustering algorithms yield diverse estimates of cluster numbers and cluster assignments. This study introduces Single Cell Consistent Clustering based on Spectral Matrix Decomposition (SCSMD), a comprehensive clustering approach that integrates the strengths of multiple methods to determine the optimal clustering scheme. Testing the performance of SCSMD across different distances and employing the bespoke evaluation metric, the methodological selection undergoes validation to ensure the optimal efficacy of the SCSMD. A consistent clustering test is conducted on 15 authentic scRNA-seq datasets. The application of SCSMD to human embryonic stem cell scRNA-seq data successfully identifies known cell types and delineates their developmental trajectories. Similarly, when applied to glioblastoma cells, SCSMD accurately detects pre-existing cell types and provides finer sub-division within one of the original clusters. The results affirm the robust performance of our SCSMD method in terms of both the number of clusters and cluster assignments. Moreover, we have broadened the application scope of SCSMD to encompass larger datasets, thereby furnishing additional evidence of its superiority. The findings suggest that SCSMD is poised for application to additional scRNA-seq datasets and for further downstream analyses.
Collapse
Affiliation(s)
- Ran Jia
- School of Mathematics and Statistics, Shandong University, Weihai 264209, Shandong, China
| | - Ying-Zan Ren
- School of Mathematics and Statistics, Shandong University, Weihai 264209, Shandong, China
| | - Po-Nian Li
- College of Mathematics and Informatics, South China Agricultural University, Guangzhou, Guangdong, China
| | - Rui Gao
- School of Control Science and Engineering, Shandong University, Jinan 250100, Shandong, China
| | - Yu-Sen Zhang
- School of Mathematics and Statistics, Shandong University, Weihai 264209, Shandong, China
| |
Collapse
|
3
|
Omotesho QA, Escamilla A, Pérez-Ruiz E, Frecha CA, Rueda-Domínguez A, Barragán I. Epigenetic targets to enhance antitumor immune response through the induction of tertiary lymphoid structures. Front Immunol 2024; 15:1348156. [PMID: 38333212 PMCID: PMC10851080 DOI: 10.3389/fimmu.2024.1348156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 01/02/2024] [Indexed: 02/10/2024] Open
Abstract
Tertiary lymphoid structures (TLS) are ectopic lymphoid aggregates found in sites of chronic inflammation such as tumors and autoimmune diseases. The discovery that TLS formation at tumor sites correlated with good patient prognosis has triggered extensive research into various techniques to induce their formation at the tumor microenvironment (TME). One strategy is the exogenous induction of specific cytokines and chemokine expression in murine models. However, applying such systemic chemokine expression can result in significant toxicity and damage to healthy tissues. Also, the TLS formed from exogenous chemokine induction is heterogeneous and different from the ones associated with favorable prognosis. Therefore, there is a need to optimize additional approaches like immune cell engineering with lentiviral transduction to improve the TLS formation in vivo. Similarly, the genetic and epigenetic regulation of the different phases of TLS neogenesis are still unknown. Understanding these molecular regulations could help identify novel targets to induce tissue-specific TLS in the TME. This review offers a unique insight into the molecular checkpoints of the different stages and mechanisms involved in TLS formation. This review also highlights potential epigenetic targets to induce TLS neogenesis. The review further explores epigenetic therapies (epi-therapy) and ongoing clinical trials using epi-therapy in cancers. In addition, it builds upon the current knowledge of tools to generate TLS and TLS phenotyping biomarkers with predictive and prognostic clinical potential.
Collapse
Affiliation(s)
- Quadri Ajibola Omotesho
- Medical Oncology Service (Group of Translational Research in Cancer Immunotherapy and Epigenetics), Regional and Clinical University Hospitals, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Malaga, Spain
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Alejandro Escamilla
- Medical Oncology Service (Group of Translational Research in Cancer Immunotherapy and Epigenetics), Regional and Clinical University Hospitals, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Malaga, Spain
- Department of Human Physiology, Human Histology, Pathological Anatomy and Physical Sport Education, University of Malaga, Malaga, Spain
| | - Elisabeth Pérez-Ruiz
- Medical Oncology Service (Group of Translational Research in Cancer Immunotherapy and Epigenetics), Regional and Clinical University Hospitals, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Malaga, Spain
| | - Cecilia A. Frecha
- Allergy Research Group, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Civil Hospital, Malaga, Spain
| | - Antonio Rueda-Domínguez
- Medical Oncology Service (Group of Translational Research in Cancer Immunotherapy and Epigenetics), Regional and Clinical University Hospitals, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Malaga, Spain
| | - Isabel Barragán
- Medical Oncology Service (Group of Translational Research in Cancer Immunotherapy and Epigenetics), Regional and Clinical University Hospitals, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Malaga, Spain
- Group of Pharmacoepigenetics, Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
4
|
Boccazzi M, Macchiarulo G, Lebon S, Janowska J, Le Charpentier T, Faivre V, Hua J, Marangon D, Lecca D, Fumagalli M, Mani S, Abbracchio MP, Gressens P, Schang AL, Van Steenwinckel J. G protein-coupled receptor 17 is regulated by WNT pathway during oligodendrocyte precursor cell differentiation. Neurobiol Dis 2023; 187:106315. [PMID: 37783234 DOI: 10.1016/j.nbd.2023.106315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 09/13/2023] [Accepted: 09/29/2023] [Indexed: 10/04/2023] Open
Abstract
G protein-coupled receptor 17 (GPR17) and the WNT pathway are critical players of oligodendrocyte (OL) differentiation acting as essential timers in developing brain to achieve fully-myelinating cells. However, whether and how these two systems are related to each other is still unknown. Of interest, both factors are dysregulated in developing and adult brain diseases, including white matter injury and cancer, making the understanding of their reciprocal interactions of potential importance for identifying new targets and strategies for myelin repair. Here, by a combined pharmacological and biotechnological approach, we examined regulatory mechanisms linking WNT signaling to GPR17 expression in OLs. We first analyzed the relative expression of mRNAs encoding for GPR17 and the T cell factor/Lymphoid enhancer-binding factor-1 (TCF/LEF) transcription factors of the canonical WNT/β-CATENIN pathway, in PDGFRα+ and O4+ OLs during mouse post-natal development. In O4+ cells, Gpr17 mRNA level peaked at post-natal day 14 and then decreased concomitantly to the physiological uprise of WNT tone, as shown by increased Lef1 mRNA level. The link between WNT signaling and GPR17 expression was further reinforced in vitro in primary PDGFRα+ cells and in Oli-neu cells. High WNT tone impaired OL differentiation and drastically reduced GPR17 mRNA and protein levels. In Oli-neu cells, WNT/β-CATENIN activation repressed Gpr17 promoter activity through both putative WNT response elements (WRE) and upregulation of the inhibitor of DNA-binding protein 2 (Id2). We conclude that the WNT pathway influences OL maturation by repressing GPR17, which could have implications in pathologies characterized by dysregulations of the OL lineage including multiple sclerosis and oligodendroglioma.
Collapse
Affiliation(s)
- Marta Boccazzi
- Université Paris Cité, Inserm, NeuroDiderot, F-75019 Paris, France; Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmaceutical Sciences, Università degli Studi di Milano, 20133 Milan, Italy
| | | | - Sophie Lebon
- Université Paris Cité, Inserm, NeuroDiderot, F-75019 Paris, France
| | - Justyna Janowska
- Université Paris Cité, Inserm, NeuroDiderot, F-75019 Paris, France; NeuroRepair Department, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | | | - Valérie Faivre
- Université Paris Cité, Inserm, NeuroDiderot, F-75019 Paris, France
| | - Jennifer Hua
- Université Paris Cité, Inserm, NeuroDiderot, F-75019 Paris, France
| | - Davide Marangon
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmaceutical Sciences, Università degli Studi di Milano, 20133 Milan, Italy
| | - Davide Lecca
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmaceutical Sciences, Università degli Studi di Milano, 20133 Milan, Italy
| | - Marta Fumagalli
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, 20133 Milan, Italy
| | - Shyamala Mani
- Université Paris Cité, Inserm, NeuroDiderot, F-75019 Paris, France
| | - Maria P Abbracchio
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmaceutical Sciences, Università degli Studi di Milano, 20133 Milan, Italy
| | - Pierre Gressens
- Université Paris Cité, Inserm, NeuroDiderot, F-75019 Paris, France
| | - Anne-Laure Schang
- Université Paris Cité, Inserm, NeuroDiderot, F-75019 Paris, France; Université Paris Cité, UMR 1153 CRESS, Paris, France.
| | | |
Collapse
|
5
|
Qiu JJ, Liu YN, Wei H, Zeng F, Yan JB. Single-cell RNA sequencing of neural stem cells derived from human trisomic iPSCs reveals the abnormalities during neural differentiation of Down syndrome. Front Mol Neurosci 2023; 16:1137123. [PMID: 37396785 PMCID: PMC10311021 DOI: 10.3389/fnmol.2023.1137123] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 05/22/2023] [Indexed: 07/04/2023] Open
Abstract
Introduction Down syndrome (DS) is the most common genetic condition that causes intellectual disability in humans. The molecular mechanisms behind the DS phenotype remain unclear. Therefore, in this study, we present new findings on its molecular mechanisms through single-cell RNA sequencing. Methods Induced pluripotent stem cells (iPSCs) from the patients with DS and the normal control (NC) patients were differentiated into iPSCs-derived neural stem cells (NSCs). Single-cell RNA sequencing was performed to achieve a comprehensive single-cell level differentiation roadmap for DS-iPSCs. Biological experiments were also performed to validate the findings. Results and Discussion The results demonstrated that iPSCs can differentiate into NSCs in both DS and NC samples. Furthermore, 19,422 cells were obtained from iPSC samples (8,500 cells for DS and 10,922 cells for the NC) and 16,506 cells from NSC samples (7,182 cells for DS and 9,324 cells for the NC), which had differentiated from the iPSCs. A cluster of DS-iPSCs, named DS-iPSCs-not differentiated (DSi-PSCs-ND), which had abnormal expression patterns compared with NC-iPSCs, were demonstrated to be unable to differentiate into DS-NSCs. Further analysis of the differentially expressed genes revealed that inhibitor of differentiation family (ID family) members, which exhibited abnormal expression patterns throughout the differentiation process from DS-iPSCs to DS-NSCs, may potentially have contributed to the neural differentiation of DS-iPSCs. Moreover, abnormal differentiation fate was observed in DS-NSCs, which resulted in the increased differentiation of glial cells, such as astrocytes, but decreased differentiation into neuronal cells. Furthermore, functional analysis demonstrated that DS-NSCs and DS-NPCs had disorders in axon and visual system development. The present study provided a new insight into the pathogenesis of DS.
Collapse
Affiliation(s)
- Jia-jun Qiu
- Shanghai Children’s Hospital, Shanghai Institute of Medical Genetics, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan-na Liu
- Shanghai Children’s Hospital, Shanghai Institute of Medical Genetics, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hao Wei
- Shanghai Children’s Hospital, Shanghai Institute of Medical Genetics, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fanyi Zeng
- Shanghai Children’s Hospital, Shanghai Institute of Medical Genetics, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Hiso-Embryology, Genetics and Developmental Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- NHC Key Laboratory of Medical Embryogenesis and Developmental Molecular Biology and Shanghai Key Laboratory of Embryo and Reproduction Engineering, Shanghai, China
| | - Jing-bin Yan
- Shanghai Children’s Hospital, Shanghai Institute of Medical Genetics, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- NHC Key Laboratory of Medical Embryogenesis and Developmental Molecular Biology and Shanghai Key Laboratory of Embryo and Reproduction Engineering, Shanghai, China
| |
Collapse
|
6
|
Wang Y, Li W, Wang M, Chen H, Li Y, Wei W, Liu X, Wu Y, Luo S, Liu X, Xiong M. Quercetin prevents the ferroptosis of OPCs by inhibiting the Id2/transferrin pathway. Chem Biol Interact 2023; 381:110556. [PMID: 37230155 DOI: 10.1016/j.cbi.2023.110556] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 05/10/2023] [Accepted: 05/17/2023] [Indexed: 05/27/2023]
Abstract
Spinal cord injury (SCI) is a destructive neurological disorder that causes impaired mobility, sensory, and autonomic dysfunctions. The loss of oligodendrocyte progenitor cells (OPCs), which can differentiate into mature oligodendrocytes and re-myelinate damaged axons, is related to poorer recovery for SCI patients. However, inhibiting OPCs loss has always been a difficult problem to overcome. In this study, we demonstrated the anti-ferroptosis effects of quercetin as a mechanism in erastin-induced OPC ferroptosis. Quercetin ameliorated erastin-induced ferroptosis in OPCs, as indicated by decreased iron concentration, reactive oxygen species (ROS) production, and increased content of glutathione (GSH) as well as more normal mitochondria morphology. Compared with erastin-induced OPCs, the myelin basic protein (MBP)-positive myelin and NF200-positive axonal was remarkably increased in quercetin-treated OPCs. Furthermore, quercetin ameliorated the erastin-induced ferroptosis as well as the myelin and axon loss of OPCs by downregulating transferrin. Transfected OPCs with transferrin overexpression plasmids significantly abrogated the protective role of quercetin in OPC ferroptosis. Using ChIP-qPCR, a direct interaction of transferrin with its upstream gene Id2 was found. The overexpression of Id2 reversed the effect of quercetin on OPC ferroptosis. In vivo study found that quercetin greatly decreased the area of injury, and enhanced the BBB score after SCI. Furthermore, in the SCI model, quercetin significantly downregulated Id2 and transferrin expression, while significantly up-regulated GPX4 and PTGS2 expression. In conclusion, quercetin prevents the ferroptosis of OPCs by inhibiting the Id2/transferrin pathway. These findings highlight quercetin as an anti-ferroptosis agent for the treatment or prevention of spinal cord injury.
Collapse
Affiliation(s)
- Yeyang Wang
- Department of Spine, Orthopaedic Center, Guangdong Second Provincial General Hospital, Guangzhou, 510000, PR China
| | - Wenjun Li
- Department of Spine, Orthopaedic Center, Guangdong Second Provincial General Hospital, Guangzhou, 510000, PR China
| | - Mingsen Wang
- Department of Orthopedic, Traditional Chinese Medicine Hospital of Puning City, Puning, 515343, PR China; Department of Orthopedic, Chaoshan Renyu Hospital of Jieyang, Jieyang, 515300, PR China
| | - Hongdong Chen
- Department of No.1 General Surgery, Guangdong Second Provincial General Hospital, Guangzhou, 510000, PR China
| | - Yongsheng Li
- Guangdong Cord Blood Bank, Guangzhou, 510663, PR China; Guangzhou Municipality Tianhe Nuoya Bio-engineering Co., Ltd, Guangzhou, 510663, PR China
| | - Wei Wei
- Guangdong Cord Blood Bank, Guangzhou, 510663, PR China; Guangzhou Municipality Tianhe Nuoya Bio-engineering Co., Ltd, Guangzhou, 510663, PR China
| | - Xuhua Liu
- Department of Orthopaedic Trauma, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510000, PR China
| | - Yuelin Wu
- Department of Spine, Orthopaedic Center, Guangdong Second Provincial General Hospital, Guangzhou, 510000, PR China
| | - Sidong Luo
- Department of Spine, Orthopaedic Center, Guangdong Second Provincial General Hospital, Guangzhou, 510000, PR China
| | - Xinfang Liu
- Department of Spine, Orthopaedic Center, Guangdong Second Provincial General Hospital, Guangzhou, 510000, PR China
| | - Man Xiong
- School of Nursing, Guangzhou University of Chinese Medicine, Guangzhou, 510000, PR China.
| |
Collapse
|
7
|
Wang X, Wu R, Zhai P, Liu Z, Xia R, Zhang Z, Qin X, Li C, Chen W, Li J, Zhang J. Hypoxia promotes EV secretion by impairing lysosomal homeostasis in HNSCC through negative regulation of ATP6V1A by HIF-1α. J Extracell Vesicles 2023; 12:e12310. [PMID: 36748335 PMCID: PMC9903130 DOI: 10.1002/jev2.12310] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 01/04/2023] [Accepted: 01/26/2023] [Indexed: 02/08/2023] Open
Abstract
Tumour cells under hypoxia tend to modulate the number and contents of extracellular vesicles (EVs) to regulate the tumour microenvironment (TME) and thus promote tumour progression. However, the mechanism of how hypoxia influences the secretion of EVs remains to be elucidated. Here, we confirm the increased production of EVs in head and neck squamous cell carcinoma (HNSCC) cells under hypoxia, where endosome-derived EVs are the main subtype affected by insufficient O2 . The accumulation of hypoxia-inducible factor-1α (HIF-1α) under hypoxia directly downregulates the expression of ATP6V1A, which is pivotal to maintain the homeostasis of lysosomes. Subsequently, impaired lysosomal degradation contributes to the reduced fusion of multivesicular bodies (MVBs) with lysosomes and enables the secretion of intraluminal vesicles (ILVs) as EVs. These findings establish a HIF-1α-regulated lysosomal dysfunction-EV release axis and provide an exquisite framework to better understand EV biogenesis.
Collapse
Affiliation(s)
- Xiaoning Wang
- Department of Oral PathologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiPRChina
- Department of Oral and Maxillofacial‐Head and Neck OncologyNinth People's Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiPRChina
| | - Ruoyi Wu
- Department of Oral and Maxillofacial‐Head and Neck OncologyNinth People's Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiPRChina
| | - Peisong Zhai
- Department of Oral and Maxillofacial‐Head and Neck OncologyNinth People's Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiPRChina
| | - Zheqi Liu
- Department of Oral and Maxillofacial‐Head and Neck OncologyNinth People's Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiPRChina
| | - Ronghui Xia
- Department of Oral PathologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiPRChina
| | - Zhen Zhang
- Department of Oral and Maxillofacial‐Head and Neck OncologyNinth People's Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiPRChina
| | - Xing Qin
- Department of Oral and Maxillofacial‐Head and Neck OncologyNinth People's Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiPRChina
| | - Chuwen Li
- Department of Oral and Maxillofacial‐Head and Neck OncologyNinth People's Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiPRChina
| | - Wantao Chen
- Department of Oral and Maxillofacial‐Head and Neck OncologyNinth People's Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiPRChina
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of StomatologyNational Clinical Research Center of StomatologyShanghaiPRChina
| | - Jiang Li
- Department of Oral PathologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiPRChina
| | - Jianjun Zhang
- Department of Oral and Maxillofacial‐Head and Neck OncologyNinth People's Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiPRChina
| |
Collapse
|
8
|
Wang Y, Yang X, Cao Y, Li X, Xu R, Yan J, Guo Z, Sun S, Sun X, Wu Y. Electroacupuncture promotes remyelination and alleviates cognitive deficit via promoting OPC differentiation in a rat model of subarachnoid hemorrhage. Metab Brain Dis 2023; 38:687-698. [PMID: 36383326 DOI: 10.1007/s11011-022-01102-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 10/10/2022] [Indexed: 11/17/2022]
Abstract
Subarachnoid hemorrhage (SAH) is a devastating cerebral vascular disease which causes neurological deficits including long-term cognitive deficit. Demyelination of white matter is correlated with cognitive deficit in SAH. Electroacupuncture (EA) is a traditional Chinese medical treatment which protects against cognitive deficit in varies of neurological diseases. However, whether EA exerts protective effect on cognitive function in SAH has not been investigated. The underlying mechanism of remyelination regulated by EA remains unclear. This study aimed to investigate the protective effects of EA on cognitive deficit in a rat model of SAH. SAH was induced in SD rats (n = 72) by endovascular perforation. Rats in EA group received EA treatment (10 min per day) under isoflurane anesthesia after SAH. Rats in SAH and sham groups received the same isoflurane anesthesia with no treatment. The mortality rate, neurological score, cognitive function, cerebral blood flow (CBF), and remyelination in sham, SAH and EA groups were assessed at 21 d after SAH.EA treatment alleviated cognitive deficits and myelin injury of rats compared with that in SAH group. Moreover, EA treatment enhanced remyelination in white matter and promoted the differentiation of OPCs after SAH. EA treatment inhibited the expression of Id2 and promoted the expression of SOX10 in oligodendrocyte cells. Additionally, the cerebral blood flow (CBF) of rats was increased by EA compared with that in SAH group. EA treatment exerts protective effect against cognitive deficit in the late phase of SAH. The underlying mechanisms involve promoting oligodendrocyte progenitor cell (OPC) differentiation and remyelination in white matter via regulating the expression of Id2 and SOX10. The improvement of CBF may also account for the protective effect of EA on cognitive function. EA treatment is a potential therapy for the treatment of cognitive deficit after SAH.
Collapse
Affiliation(s)
- Yingwen Wang
- Departement of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, NO.1 of Youyi Road, Yuzhong District, Chongqing, China
| | - Xiaomin Yang
- Departement of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, NO.1 of Youyi Road, Yuzhong District, Chongqing, China
| | - Yunchuan Cao
- Departement of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, NO.1 of Youyi Road, Yuzhong District, Chongqing, China
| | - Xiaoguo Li
- Departement of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, NO.1 of Youyi Road, Yuzhong District, Chongqing, China
| | - Rui Xu
- Departement of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, NO.1 of Youyi Road, Yuzhong District, Chongqing, China
| | - Jin Yan
- Departement of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, NO.1 of Youyi Road, Yuzhong District, Chongqing, China
| | - Zongduo Guo
- Departement of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, NO.1 of Youyi Road, Yuzhong District, Chongqing, China
| | - Shanquan Sun
- Institute of Neuroscience, Chongqing Medical University, Chongqing, China
| | - Xiaochuan Sun
- Departement of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, NO.1 of Youyi Road, Yuzhong District, Chongqing, China.
| | - Yue Wu
- Departement of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, NO.1 of Youyi Road, Yuzhong District, Chongqing, China.
| |
Collapse
|
9
|
Su X, Ying J, Xiao D, Qiu X, Li S, Zhao F, Tang J. Activin A rescues preterm brain injury through a novel Noggin/BMP4/Id2 signaling pathway. Int J Mol Med 2022; 51:12. [PMID: 36524372 PMCID: PMC9848437 DOI: 10.3892/ijmm.2022.5215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 12/07/2022] [Indexed: 12/15/2022] Open
Abstract
Activin A (Act A) has been reported to promote oligodendrocyte progenitor cell (OPC) differentiation in vitro and improve neurological outcomes in adult mice. However, the roles and mechanisms of action of Act A in preterm brain injury are unknown. In the present study, P5 rats were subjected to hypoxia‑ischemia to establish a neonatal white matter injury (WMI) model and Act A was injected via the lateral ventricle. Pathological characteristics, OPC differentiation, myelination, and neurological performance were analyzed. Further, the involvement of the Noggin/BMP4/Id2 signaling pathway in the roles of Act A in WMI was explored. Act A attenuated pathological damage, promoted OPC differentiation, enhanced myelin sheath and myelinated axon formation, and improved neurological performance of WMI rats. Moreover, Act A enhanced noggin expression, which, in turn, inhibited the expression of bone morphogenetic protein 4 (BMP4) and inhibitor of DNA binding 2 (Id2). Furthermore, upregulation of Id2 completely abolished the rescue effects of Act A in WMI rats. In conclusion, the present findings suggested that Act A rescues preterm brain injury via targeting a novel Noggin/BMP4/Id2 signaling pathway.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jun Tang
- Correspondence to: Professor Jun Tang, Department of Pediatrics/Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University Hospital, Sichuan University, Section 3, 20 South Renmin Road, Chengdu, Sichuan 610041, P.R. China, E-mail:
| |
Collapse
|
10
|
Hippocampal miR-211-5p regulates neurogenesis and depression-like behaviors in the rat. Neuropharmacology 2021; 194:108618. [PMID: 34062164 DOI: 10.1016/j.neuropharm.2021.108618] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 05/13/2021] [Accepted: 05/15/2021] [Indexed: 01/22/2023]
Abstract
Emerging evidence has shown that microRNAs (miRNAs) contribute to the pathogenesis of depression, a potentially life-threatening and disabling mental disorder caused by the interaction of genetic and environmental factors. However, the specific miRNAs and their underlying molecular mechanisms as involved in the pathogenesis and development of depression remain largely unknown. In the present study, we screened miRNA expression profiles and found that miR-211-5p was significantly down-regulated within the dentate gyrus (DG) hippocampus in the chronic unpredictable mild stress (CUMS) induced rat model of depression. Deficits in miR-211-5p were accompanied with reductions in neurogenesis and increased apoptosis in these CUMS rats. In contrast, an up-regulation of miR-211-5p within the DG area in CUMS rats promoted neuronal neurogenesis, reduced neuronal apoptosis via suppression of the Dyrk1A/STAT3 signaling pathway and relieved depression-like behaviors in these CUMS rats. In rats subjected to a knock-down of miR-211-5p in the DG there was an increase in neuronal apoptosis and a decrease in neuronal regeneration, effects which were accompanied with an induction of depression-like behaviors. Taken together, the results of our study reveal that altered levels of miR-211-5p in the hippocampal DG area exert a significant impact on neurogenesis, apoptosis and thus depression-like behaviors in rats. These findings suggest that the miR-211-5p/Dyrk1A pathway plays an important role in the pathogenesis of depression and may serve as a potential therapeutic target for the treatment of depression.
Collapse
|
11
|
Li J, Shang Y, Wang L, Zhao B, Sun C, Li J, Liu S, Li C, Tang M, Meng FL, Zheng P. Genome integrity and neurogenesis of postnatal hippocampal neural stem/progenitor cells require a unique regulator Filia. SCIENCE ADVANCES 2020; 6:6/44/eaba0682. [PMID: 33115731 PMCID: PMC7608785 DOI: 10.1126/sciadv.aba0682] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 09/01/2020] [Indexed: 05/03/2023]
Abstract
Endogenous DNA double-strand breaks (DSBs) formation and repair in neural stem/progenitor cells (NSPCs) play fundamental roles in neurogenesis and neurodevelopmental disorders. NSPCs exhibit heterogeneity in terms of lineage fates and neurogenesis activity. Whether NSPCs also have heterogeneous regulations on DSB formation and repair to accommodate region-specific neurogenesis has not been explored. Here, we identified a regional regulator Filia, which is predominantly expressed in mouse hippocampal NSPCs after birth and regulates DNA DSB formation and repair. On one hand, Filia protects stalling replication forks and prevents the replication stress-associated DNA DSB formation. On the other hand, Filia facilitates the homologous recombination-mediated DNA DSB repair. Consequently, Filia-/- mice had impaired hippocampal NSPC proliferation and neurogenesis and were deficient in learning, memory, and mood regulations. Thus, our study provided the first proof of concept demonstrating the region-specific regulations of DSB formation and repair in subtypes of NSPCs.
Collapse
Affiliation(s)
- Jingzheng Li
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- Yunnan Key Laboratory of Animal Reproduction, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Yafang Shang
- University of Chinese Academy of Sciences, Beijing 101408, China
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Lin Wang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- Yunnan Key Laboratory of Animal Reproduction, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Bo Zhao
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- Yunnan Key Laboratory of Animal Reproduction, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Chunli Sun
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- Yunnan Key Laboratory of Animal Reproduction, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Jiali Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan 650201, China
| | - Siling Liu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan 650201, China
| | - Cong Li
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- Yunnan Key Laboratory of Animal Reproduction, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Min Tang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- Yunnan Key Laboratory of Animal Reproduction, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Fei-Long Meng
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China.
| | - Ping Zheng
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China.
- Yunnan Key Laboratory of Animal Reproduction, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming 650201, China
| |
Collapse
|
12
|
Genetic Base of Behavioral Disorders in Mucopolysaccharidoses: Transcriptomic Studies. Int J Mol Sci 2020; 21:ijms21031156. [PMID: 32050523 PMCID: PMC7036933 DOI: 10.3390/ijms21031156] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 01/31/2020] [Accepted: 02/06/2020] [Indexed: 12/15/2022] Open
Abstract
Mucopolysaccharidoses (MPS) are a group of inherited metabolic diseases caused by mutations leading to defective degradation of glycosaminoglycans (GAGs) and their accumulation in cells. Among 11 known types and subtypes of MPS, neuronopathy occurs in seven (MPS I, II, IIIA, IIIB, IIIC, IIID, VII). Brain dysfunctions, occurring in these seven types/subtypes include various behavioral disorders. Intriguingly, behavioral symptoms are significantly different between patients suffering from various MPS types. Molecular base of such differences remains unknown. Here, we asked if expression of genes considered as connected to behavior (based on Gene Ontology, GO terms) is changed in MPS. Using cell lines of all MPS types, we have performed transcriptomic (RNA-seq) studies and assessed expression of genes involved in behavior. We found significant differences between MPS types in this regard, with the most severe changes in MPS IIIA (the type considered as the behaviorally most severely affected), while the lowest changes in MPS IVA and MPS VI (types in which little or no behavioral disorders are known). Intriguingly, relatively severe changes were found also in MPS IVB (in which, despite no behavioral disorder noted, the same gene is mutated as in GM1 gangliosidosis, a severe neurodegenerative disease) and MPS IX (in which only a few patients were described to date, thus, behavioral problems are not well recognized). More detailed analyses of expression of certain genes allowed us to propose an association of specific changes in the levels of transcripts in specific MPS types to certain behavioral disorders observed in patients. Therefore, this work provides a principle for further studies on the molecular mechanism of behavioral changes occurring in MPS patients.
Collapse
|
13
|
Ornelas IM, Khandker L, Wahl SE, Hashimoto H, Macklin WB, Wood TL. The mechanistic target of rapamycin pathway downregulates bone morphogenetic protein signaling to promote oligodendrocyte differentiation. Glia 2020; 68:1274-1290. [PMID: 31904150 DOI: 10.1002/glia.23776] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 11/26/2019] [Accepted: 11/29/2019] [Indexed: 12/13/2022]
Abstract
Oligodendrocyte precursor cells (OPCs) differentiate and mature into oligodendrocytes, which produce myelin in the central nervous system. Prior studies have shown that the mechanistic target of rapamycin (mTOR) is necessary for proper myelination of the mouse spinal cord and that bone morphogenetic protein (BMP) signaling inhibits oligodendrocyte differentiation, in part by promoting expression of inhibitor of DNA binding 2 (Id2). Here we provide evidence that mTOR functions specifically in the transition from early stage OPC to immature oligodendrocyte by downregulating BMP signaling during postnatal spinal cord development. When mTOR is deleted from the oligodendrocyte lineage, expression of the FK506 binding protein 1A (FKBP12), a suppressor of BMP receptor activity, is reduced, downstream Smad activity is increased and Id2 expression is elevated. Additionally, mTOR inhibition with rapamycin in differentiating OPCs alters the transcriptional complex present at the Id2 promoter. Deletion of mTOR in oligodendroglia in vivo resulted in fewer late stage OPCs and fewer newly formed oligodendrocytes in the spinal cord with no effect on OPC proliferation or cell cycle exit. Finally, we demonstrate that inhibiting BMP signaling rescues the rapamycin-induced deficit in myelin protein expression. We conclude that mTOR promotes early oligodendrocyte differentiation by suppressing BMP signaling in OPCs.
Collapse
Affiliation(s)
- Isis M Ornelas
- Department of Pharmacology, Physiology & Neuroscience, New Jersey Medical School, Rutgers University, Newark, New Jersey
| | - Luipa Khandker
- Department of Pharmacology, Physiology & Neuroscience, New Jersey Medical School, Rutgers University, Newark, New Jersey
| | - Stacey E Wahl
- Department of Pharmacology, Physiology & Neuroscience, New Jersey Medical School, Rutgers University, Newark, New Jersey
| | - Hirokazu Hashimoto
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, Colorado
| | - Wendy B Macklin
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, Colorado
| | - Teresa L Wood
- Department of Pharmacology, Physiology & Neuroscience, New Jersey Medical School, Rutgers University, Newark, New Jersey
| |
Collapse
|
14
|
Li F, Liu WC, Wang Q, Sun Y, Wang H, Jin X. NG2-glia cell proliferation and differentiation by glial growth factor 2 (GGF2), a strategy to promote functional recovery after ischemic stroke. Biochem Pharmacol 2019; 171:113720. [PMID: 31751533 DOI: 10.1016/j.bcp.2019.113720] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Accepted: 11/14/2019] [Indexed: 12/16/2022]
Abstract
Stroke is the leading cause of adult disability. Spontaneous functional recovery occurs after ischemic stroke, but it is very limited. Therefore, it is urgent to find a strategy to promote functional recovery after stroke in clinical setting. Gray matter damage has received extensive attention owing to the important roles of the gray matter in synaptic plasticity, cognitive, and motor function. However, stroke also causes white matter damage, which accounts for half of the infarct volume and can be aggravated by blood brain barrier damage. Disruption of white matter integrity, which is characterized by death of oligodendrocytes (OLs), loss of myelin, and axonal injury, greatly contributes to impaired neurological function. Impaired proliferation and differentiation of OL precursor cell (OPC, NG2-glia cells) play an important role in limited functional recovery after ischemic stroke and inhibitor of differentiation 2 (ID2) is a key factor controlling NG2-glia cells differentiation. It has been reported that the number of NG2-glia cells in the peri-infarction area significantly increases after ischemic stroke and glial growth factor (GGF2) administration promotes the proliferation and differentiation of NG2-glia cells as well as functional recovery after spinal cord injury. On the basis of the important roles of GGF2 in functional recovery and those of ID2 in NG2-glia cell proliferation and differentiation, we propose that after binding with the ErBb receptor on the surface of NG2-glia cells, GGF2 promotes NG2-glia cell proliferation and differentiation, thereby repairing BBB and white matter integrity and promoting neural functional recovery after ischemic stroke.
Collapse
Affiliation(s)
- Fei Li
- School of Pharmaceutical Sciences, Hubei University of Medicine, Shiyan 442000, China; Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan 442000, China
| | - Wen-Cao Liu
- Shanxi Provincial People's Hospital, Taiyuan 030001, China
| | - Qi Wang
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Yanyun Sun
- Jiangsu Key Laboratory of Neuro-Psychiatry Research and Institute of Neuroscience, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China.
| | - Hongbo Wang
- Key Laboratory of Molecular Pharmacology and Drug Evaluation, School of Pharmacy, Yantai University, Yantai, China.
| | - Xinchun Jin
- Jiangsu Key Laboratory of Neuro-Psychiatry Research and Institute of Neuroscience, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China; Key Laboratory of Molecular Pharmacology and Drug Evaluation, School of Pharmacy, Yantai University, Yantai, China.
| |
Collapse
|
15
|
Jiménez-González V, Ogalla-García E, García-Quintanilla M, García-Quintanilla A. Deciphering GRINA/Lifeguard1: Nuclear Location, Ca 2+ Homeostasis and Vesicle Transport. Int J Mol Sci 2019; 20:ijms20164005. [PMID: 31426446 PMCID: PMC6719933 DOI: 10.3390/ijms20164005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 07/31/2019] [Accepted: 08/12/2019] [Indexed: 01/31/2023] Open
Abstract
The Glutamate Receptor Ionotropic NMDA-Associated Protein 1 (GRINA) belongs to the Lifeguard family and is involved in calcium homeostasis, which governs key processes, such as cell survival or the release of neurotransmitters. GRINA is mainly associated with membranes of the endoplasmic reticulum, Golgi, endosome, and the cell surface, but its presence in the nucleus has not been explained yet. Here we dissect, with the help of different software tools, the potential roles of GRINA in the cell and how they may be altered in diseases, such as schizophrenia or celiac disease. We describe for the first time that the cytoplasmic N-terminal half of GRINA (which spans a Proline-rich domain) contains a potential DNA-binding sequence, in addition to cleavage target sites and probable PY-nuclear localization sequences, that may enable it to be released from the rest of the protein and enter the nucleus under suitable conditions, where it could participate in the transcription, alternative splicing, and mRNA export of a subset of genes likely involved in lipid and sterol synthesis, ribosome biogenesis, or cell cycle progression. To support these findings, we include additional evidence based on an exhaustive review of the literature and our preliminary data of the protein–protein interaction network of GRINA.
Collapse
Affiliation(s)
| | - Elena Ogalla-García
- Department of Pharmacology, School of Pharmacy, University of Seville, 41012 Seville, Spain
| | - Meritxell García-Quintanilla
- Institute of Biomedicine of Seville (IBiS), University Hospital Virgen del Rocío/CSIC/University of Seville, 41013 Seville, Spain
| | - Albert García-Quintanilla
- Department of Biochemistry and Molecular Biology, School of Pharmacy, University of Seville, 41012 Seville, Spain.
| |
Collapse
|
16
|
Kang M, Yao Y. Oligodendrocytes in intracerebral hemorrhage. CNS Neurosci Ther 2019; 25:1075-1084. [PMID: 31410988 PMCID: PMC6776757 DOI: 10.1111/cns.13193] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 06/23/2019] [Accepted: 06/26/2019] [Indexed: 12/22/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is a cerebrovascular disorder with high mortality and disability rates. Although a lot of effort has been put in ICH, there is still no effective treatment for this devastating disease. Recent studies suggest that oligodendrocytes play an important role in brain repair after ICH and thus may be targeted for the therapies of ICH. Here in this review, we first introduce the origin, migration, proliferation, differentiation, and myelination of oligodendrocytes under physiological condition. Second, recent findings on how ICH affects oligodendrocyte biology and function are reviewed. Third, potential crosstalk between oligodendrocytes and other cells in the brain is also summarized. Last, we discuss the therapeutic potential of oligodendrocyte‐based treatments in ICH. Our goal is to provide a comprehensive review on the biology and function of oligodendrocytes under both physiological and ICH conditions.
Collapse
Affiliation(s)
- Minkyung Kang
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, GA, USA
| | - Yao Yao
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, GA, USA
| |
Collapse
|
17
|
APC-Cdh1 Inhibits the Proliferation and Activation of Oligodendrocyte Precursor Cells after Mechanical Stretch Injury. BIOMED RESEARCH INTERNATIONAL 2019; 2019:9524561. [PMID: 31139661 PMCID: PMC6500630 DOI: 10.1155/2019/9524561] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 03/25/2019] [Indexed: 12/21/2022]
Abstract
The incidence of spinal cord injury (SCI) continues to increase; however, the involved mechanisms remain unclear. Anaphase promoting complex (APC) and its regulatory subunit Cdh1 play important roles in the growth, development, and repair of the central nervous system (CNS). Cdh1 is involved in the pathophysiological processes of neuronal apoptosis and astrocyte-reactive proliferation after ischemic brain injury, whereas the role played by APC-Cdh1 in the proliferation and activation of oligodendrocyte precursor cells (OPCs) after SCI remains unresolved. Using primary cultures of spinal oligodendrocyte precursor cells, we successfully established an in vitro mechanical stretch injury model to simulate SCI. Cell viability and proliferation were determined by MTT assay and flow cytometric analysis of the cell cycle. Real-time fluorescent quantitative PCR and Western blot analysis determined the mRNA and protein expression levels of Cdh1 and its downstream substrates Skp2 and Id2. Mechanical stretch injury decreased the proliferative activity of OPCs and enhanced cellular Cdh1 expression. Dampened expression of Cdh1 in primary OPCs significantly promoted proliferation and activation of OPCs after SCI. In addition, the expression of the downstream substrates of Cdh1, Skp2, and Id2 was decreased following mechanical injury, whereas adenovirus-mediated Cdh1 RNA interference increased postinjury expression of Skp2 and Id2. These findings suggest that APC-Cdh1 might be involved in regulating the proliferation and activation of OPCs after mechanical SCI. Moreover, degraded ubiquitination of the downstream substrates Skp2 and Id2 might play an important role, at least in part, in the beneficial effects of OPCs activity following SCI.
Collapse
|
18
|
Zhou XL, Zeng D, Ye YH, Sun SM, Lu XF, Liang WQ, Chen CF, Lin HY. Prognostic values of the inhibitor of DNA‑binding family members in breast cancer. Oncol Rep 2018; 40:1897-1906. [PMID: 30066902 PMCID: PMC6111598 DOI: 10.3892/or.2018.6589] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 07/17/2018] [Indexed: 02/05/2023] Open
Abstract
The inhibitor of DNA‑binding (ID) proteins are dominant‑negative modulators of transcription factors with basic helix‑loop‑helix (bHLH) structures, which control a variety of genes in cell cycle regulation. An increasing volume of evidence has demonstrated that the deregulated expression of IDs in several types of malignancy, including breast carcinoma, has been proven to serve crucial regulatory functions in tumorigenesis and the development of breast cancer (BC). The present study evaluated the prognostic values of the ID family members by investigating a set of publicly accessible databases, including Oncomine, bc‑GenExMiner, Kaplan‑Meier plotter and the Human Protein Atlas. The results demonstrated that mRNA levels of distinct IDs exhibited diverse profiles between BC and normal counterparts. The mRNA expression level of ID2 was significantly higher in breast cancer than normal tissues, while the mRNA expression levels of ID1, ID3 and ID4 were significantly lower in breast cancer tissues than in normal tissues. Furthermore, higher mRNA expression levels of ID1 and ID4 were associated with subgroups with lower pathological grades and fewer lymph node metastases. Survival analysis revealed that elevated mRNA levels of ID1 and ID4 predicted an improved survival in all patients with BC. Increased ID1 mRNA levels were associated with higher relapse‑free survival rates in all patients with BC, particularly in those with ER positive and Luminal A subtype tumors. Increased ID4 mRNA expression predicted longer survival times in all patients with BC, particularly in those with hormone receptor‑positive tumors or those treated with endocrine therapy. These results indicated that IDs are essential prognostic indicators in BC. Future studies on the effect of IDs on the pathogenesis and development of BC are warranted.
Collapse
Affiliation(s)
- Xiao-Ling Zhou
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515000, P.R. China
| | - De Zeng
- Department of Medical Oncology, The Cancer Hospital of Shantou University Medical College, Shantou, Guangdong 515000, P.R. China
| | - Yan-Hong Ye
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515000, P.R. China
| | - Shu-Ming Sun
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515000, P.R. China
| | - Xiao-Feng Lu
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515000, P.R. China
| | - Wei-Quan Liang
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515000, P.R. China
| | - Chun-Fa Chen
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515000, P.R. China
| | - Hao-Yu Lin
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515000, P.R. China
- Correspondence to: Dr Hao-Yu Lin, Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Shantou University Medical College (SUMC), 57 Changping Road, Shantou, Guangdong 515000, P.R. China, E-mail:
| |
Collapse
|
19
|
Wen XF, Chen M, Wu Y, Chen MN, Glogowska A, Klonisch T, Zhang GJ. Inhibitor of DNA Binding 2 Inhibits Epithelial-Mesenchymal Transition via Up-Regulation of Notch3 in Breast Cancer. Transl Oncol 2018; 11:1259-1270. [PMID: 30119050 PMCID: PMC6097462 DOI: 10.1016/j.tranon.2018.07.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 07/23/2018] [Indexed: 02/05/2023] Open
Abstract
Breast cancer is the second leading cause of cancer death in women worldwide. Incurable metastatic breast disease presents a major clinical challenge and is the main cause of breast cancer-related death. The epithelial-mesenchymal transition (EMT) is a critical early promoter of metastasis. In the present study, we identified a novel role for the inhibitor of DNA binding 2 (Id2), a member of the basic helix-loop-helix protein family, during the EMT of breast cancer. Expression of Id2 was positively correlated with Notch3 in breast cancer cells. Low expression of Id2 and Notch3 was associated with worse distant metastasis-free survival in breast cancer patients. The present study revealed that Id2 activated Notch3 expression by blocking E2A binding to an E-box motif in the Notch3 promoter. The Id2-mediated up-regulation of Notch3 expression at both the mRNA and protein levels resulted in an attenuated EMT, which was associated with reduced motility and matrix invasion of ER-positive and -negative human breast cancer cells and the emergence of E-cadherin expression and reduction in the mesenchymal marker vimentin in triple-negative breast cancer cells. In summary, our findings identified Id2 as a suppressor of the EMT and positive transcriptional regulator of Notch3 in breast cancer. Id2 and Notch3 may serve as novel prognostic markers in a subpopulation of ER-positive breast cancer patients.
Collapse
Affiliation(s)
- Xiao-Fen Wen
- Department of Breast Medical Oncology, Cancer Hospital of Shantou University Medical College, 7 Raoping Road, Shantou, China; ChangJiang Scholar's Laboratory, Shantou University Medical College, 22 Xinling Road, Shantou, China
| | - Min Chen
- ChangJiang Scholar's Laboratory, Shantou University Medical College, 22 Xinling Road, Shantou, China; Xiang'an Hospital, Xiamen University, 2000 East Xiang'an Rd, Xiamen, Fujian, China
| | - Yang Wu
- ChangJiang Scholar's Laboratory, Shantou University Medical College, 22 Xinling Road, Shantou, China
| | - Min-Na Chen
- Department of Breast Medical Oncology, Cancer Hospital of Shantou University Medical College, 7 Raoping Road, Shantou, China; ChangJiang Scholar's Laboratory, Shantou University Medical College, 22 Xinling Road, Shantou, China
| | - Aleksandra Glogowska
- Dept. of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
| | - Thomas Klonisch
- Dept. of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada.
| | - Guo-Jun Zhang
- ChangJiang Scholar's Laboratory, Shantou University Medical College, 22 Xinling Road, Shantou, China; Xiang'an Hospital, Xiamen University, 2000 East Xiang'an Rd, Xiamen, Fujian, China.
| |
Collapse
|
20
|
Sustained Expression of Negative Regulators of Myelination Protects Schwann Cells from Dysmyelination in a Charcot-Marie-Tooth 1B Mouse Model. J Neurosci 2018; 38:4275-4287. [PMID: 29610440 DOI: 10.1523/jneurosci.0201-18.2018] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 03/02/2018] [Accepted: 03/16/2018] [Indexed: 12/16/2022] Open
Abstract
Schwann cell differentiation and myelination in the PNS are the result of fine-tuning of positive and negative transcriptional regulators. As myelination starts, negative regulators are downregulated, whereas positive ones are upregulated. Fully differentiated Schwann cells maintain an extraordinary plasticity and can transdifferentiate into "repair" Schwann cells after nerve injury. Reactivation of negative regulators of myelination is essential to generate repair Schwann cells. Negative regulators have also been implicated in demyelinating neuropathies, although their role in disease remains elusive. Here, we used a mouse model of Charcot-Marie-Tooth neuropathy type 1B (CMT1B), the P0S63del mouse characterized by ER stress and the activation of the unfolded protein response, to show that adult Schwann cells are in a partial differentiation state because they overexpress transcription factors that are normally expressed only before myelination. We provide evidence that two of these factors, Sox2 and Id2, act as negative regulators of myelination in vivo However, their sustained expression in neuropathy is protective because ablation of Sox2 or/and Id2 from S63del mice of both sexes results in worsening of the dysmyelinating phenotype. This is accompanied by increased levels of mutant P0 expression and exacerbation of ER stress, suggesting that limited differentiation may represent a novel adaptive mechanism through which Schwann cells counter the toxic effect of a mutant terminal differentiation protein.SIGNIFICANCE STATEMENT In many neuropathies, Schwann cells express high levels of early differentiation genes, but the significance of these altered expression remained unclear. Because many of these factors may act as negative regulators of myelination, it was suggested that their misexpression could contribute to dysmyelination. Here, we show that the transcription factors Sox2 and Id2 act as negative regulators of myelination in vivo, but that their sustained expression in Charcot-Marie-Tooth type 1B (CMT1B) represents an adaptive response activated by the Schwann cells to reduce mutant protein toxicity and prevent demyelination.
Collapse
|
21
|
Wang CY, Deneen B, Tzeng SF. MicroRNA-212 inhibits oligodendrocytes during maturation by down-regulation of differentiation-associated gene expression. J Neurochem 2017; 143:112-125. [DOI: 10.1111/jnc.14138] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2017] [Revised: 07/22/2017] [Accepted: 07/31/2017] [Indexed: 02/06/2023]
Affiliation(s)
- Chih-Yen Wang
- Department of Life Sciences; College of Bioscience and Biotechnology; National Cheng Kung University; Tainan Taiwan
| | - Benjamin Deneen
- Center for Cell and Gene Therapy; Baylor College of Medicine; Houston Texas USA
| | - Shun-Fen Tzeng
- Department of Life Sciences; College of Bioscience and Biotechnology; National Cheng Kung University; Tainan Taiwan
| |
Collapse
|
22
|
Rossi M, Spichty M, Attorri L, Distante C, Nervi C, Salvati S, Vitelli L. Eicosapentaenoic acid modulates the synergistic action of CREB1 and ID/E2A family members in the rat pup brain and mouse embryonic stem cells. BIOCHIMICA ET BIOPHYSICA ACTA. GENE REGULATORY MECHANISMS 2017; 1860:870-884. [PMID: 28666847 DOI: 10.1016/j.bbagrm.2017.06.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 06/01/2017] [Accepted: 06/20/2017] [Indexed: 12/13/2022]
Abstract
The aim of this study was to investigate the molecular mechanism by which eicosapentaenoic acid (EPA) may exert neuroprotective effects through an "EPA-cyclic AMP response element-binding protein (CREB)" signaling pathway. The current study reveals that EPA modulates the exquisite interplay of interaction of CREB1 with the inhibitor of DNA binding (ID) and E2A family members, thereby delivering mechanistic insights into specific neural differentiation program. In this scenario, our work provides evidence for the capability of CREB1 to sequester ID:E2A family members in brain tissues and neural differentiating mouse embryonic stem cells (mESCs) through formation of a [CREB1]2:ID2:E47 tetrameric complex.In essence, the molecular function of CREB1 is to dynamically regulate the location-specific assembly or disassembly of basic-helix-loop-helix (bHLH):HLH protein complexes to mediate the activation of neural/glial target genes. Together, these findings support the one-to-many binding mechanism of CREB1 and indicate that EPA treatment potentiates the integration of CREB dependent signaling with HLH/bHLH transcriptional network, adding specificity to the CREB1-mediated gene regulation during neural/glial differentiation. Our current research on the EPA-CREB axis could reveal new molecular targets for treating neurogenerative disease.
Collapse
Affiliation(s)
- Maurizio Rossi
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Martin Spichty
- Laboratory of Biology and Modelling of the Cell, Lyon University, ENS Lyon, University Claude Bernard, CNRS UMR 5239, INSERM U1210, 46 allée d'Italie, Site Jacques Monod, F-69007 Lyon, France
| | - Lucilla Attorri
- Department of Public Veterinary Health and Food Safety, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Chiara Distante
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Clara Nervi
- Department of Medical and Surgical Sciences and Biotechnologies, University of Roma "La Sapienza", Corso della Repubblica 79, 04100, Latina, Italy
| | - Serafina Salvati
- Department of Public Veterinary Health and Food Safety, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Luigi Vitelli
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy.
| |
Collapse
|
23
|
Tan BT, Jiang L, Liu L, Yin Y, Luo ZRX, Long ZY, Li S, Yu LH, Wu YM, Liu Y. Local injection of Lenti-Olig2 at lesion site promotes functional recovery of spinal cord injury in rats. CNS Neurosci Ther 2017; 23:475-487. [PMID: 28452182 DOI: 10.1111/cns.12694] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 03/08/2017] [Accepted: 03/10/2017] [Indexed: 12/17/2022] Open
Abstract
AIMS Olig2 is one of the most critical factors during CNS development, which belongs to b-HLH transcription factor family. Previous reports have shown that Olig2 regulates the remyelination processes in CNS demyelination diseases models. However, the role of Olig2 in contusion spinal cord injury (SCI) and the possible therapeutic effects remain obscure. This study aims to investigate the effects of overexpression Olig2 by lentivirus on adult spinal cord injury rats. METHODS Lenti-Olig2 expression and control Lenti-eGFP vectors were prepared, and virus in a total of 5 μL (108 TU/mL) was locally injected into the injured spinal cord 1.5 mm rostral and caudal near the epicenter. Immunostaining, Western blot, electron microscopy, and CatWalk analyzes were employed to investigate the effects of Olig2 on spinal cord tissue repair and functional recovery. RESULTS Injection of Lenti-Olig2 significantly increased the number of oligodendrocytes lineage cells and enhanced myelination after SCI. More importantly, the introduction of Olig2 greatly improved hindlimb locomotor performances. Other oligodendrocyte-related transcription factors, which were downregulated or upregulated after injury, were reversed by Olig2 induction. CONCLUSIONS Our findings provided the evidence that overexpression Olig2 promotes myelination and locomotor recovery of contusion SCI, which gives us more understanding of Olig2 on spinal cord injury treatment.
Collapse
Affiliation(s)
- Bo-Tao Tan
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Research Institute of Surgery, Daping Hospital, The Third Military Medical University, Chongqing, China.,Department of Rehabilitation Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Long Jiang
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Research Institute of Surgery, Daping Hospital, The Third Military Medical University, Chongqing, China
| | - Li Liu
- Department of Brain, The Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Ying Yin
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ze-Ru-Xin Luo
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zai-Yun Long
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Research Institute of Surgery, Daping Hospital, The Third Military Medical University, Chongqing, China
| | - Sen Li
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Research Institute of Surgery, Daping Hospital, The Third Military Medical University, Chongqing, China
| | - Le-Hua Yu
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ya-Min Wu
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Research Institute of Surgery, Daping Hospital, The Third Military Medical University, Chongqing, China
| | - Yuan Liu
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Research Institute of Surgery, Daping Hospital, The Third Military Medical University, Chongqing, China
| |
Collapse
|
24
|
Abstract
Inhibitors of DNA binding and cell differentiation (Id) proteins are members of the large family of the helix-loop-helix (HLH) transcription factors, but they lack any DNA-binding motif. During development, the Id proteins play a key role in the regulation of cell-cycle progression and cell differentiation by modulating different cell-cycle regulators both by direct and indirect mechanisms. Several Id-protein interacting partners have been identified thus far, which belong to structurally and functionally unrelated families, including, among others, the class I and II bHLH transcription factors, the retinoblastoma protein and related pocket proteins, the paired-box transcription factors, and the S5a subunit of the 26 S proteasome. Although the HLH domain of the Id proteins is involved in most of their protein-protein interaction events, additional motifs located in their N-terminal and C-terminal regions are required for the recognition of diverse protein partners. The ability of the Id proteins to interact with structurally different proteins is likely to arise from their conformational flexibility: indeed, these proteins contain intrinsically disordered regions that, in the case of the HLH region, undergo folding upon self- or heteroassociation. Besides their crucial role for cell-fate determination and cell-cycle progression during development, other important cellular events have been related to the Id-protein expression in a number of pathologies. Dysregulated Id-protein expression has been associated with tumor growth, vascularization, invasiveness, metastasis, chemoresistance and stemness, as well as with various developmental defects and diseases. Herein we provide an overview on the structural properties, mode of action, biological function and therapeutic potential of these regulatory proteins.
Collapse
Affiliation(s)
- Cornelia Roschger
- Department of Molecular Biology, University of Salzburg, Billrothstrasse 11, Salzburg, 5020, Austria
| | - Chiara Cabrele
- Department of Molecular Biology, University of Salzburg, Billrothstrasse 11, Salzburg, 5020, Austria.
| |
Collapse
|
25
|
Roschger C, Cabrele C. The Id-protein family in developmental and cancer-associated pathways. Cell Commun Signal 2017; 15:7. [PMID: 28122577 PMCID: PMC5267474 DOI: 10.1186/s12964-016-0161-y] [Citation(s) in RCA: 135] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 12/29/2016] [Indexed: 01/15/2023] Open
Abstract
Inhibitors of DNA binding and cell differentiation (Id) proteins are members of the large family of the helix-loop-helix (HLH) transcription factors, but they lack any DNA-binding motif. During development, the Id proteins play a key role in the regulation of cell-cycle progression and cell differentiation by modulating different cell-cycle regulators both by direct and indirect mechanisms. Several Id-protein interacting partners have been identified thus far, which belong to structurally and functionally unrelated families, including, among others, the class I and II bHLH transcription factors, the retinoblastoma protein and related pocket proteins, the paired-box transcription factors, and the S5a subunit of the 26 S proteasome. Although the HLH domain of the Id proteins is involved in most of their protein-protein interaction events, additional motifs located in their N-terminal and C-terminal regions are required for the recognition of diverse protein partners. The ability of the Id proteins to interact with structurally different proteins is likely to arise from their conformational flexibility: indeed, these proteins contain intrinsically disordered regions that, in the case of the HLH region, undergo folding upon self- or heteroassociation. Besides their crucial role for cell-fate determination and cell-cycle progression during development, other important cellular events have been related to the Id-protein expression in a number of pathologies. Dysregulated Id-protein expression has been associated with tumor growth, vascularization, invasiveness, metastasis, chemoresistance and stemness, as well as with various developmental defects and diseases. Herein we provide an overview on the structural properties, mode of action, biological function and therapeutic potential of these regulatory proteins.
Collapse
Affiliation(s)
- Cornelia Roschger
- Department of Molecular Biology, University of Salzburg, Billrothstrasse 11, Salzburg, 5020, Austria
| | - Chiara Cabrele
- Department of Molecular Biology, University of Salzburg, Billrothstrasse 11, Salzburg, 5020, Austria.
| |
Collapse
|
26
|
Kim SY, Kelland EE, Kim JH, Lund BT, Chang X, Wang K, Weiner LP. The influence of retinoic acid on the human oligodendrocyte precursor cells by RNA-sequencing. Biochem Biophys Rep 2016; 9:166-172. [PMID: 29114585 PMCID: PMC5632710 DOI: 10.1016/j.bbrep.2016.12.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 11/01/2016] [Accepted: 12/13/2016] [Indexed: 11/16/2022] Open
Abstract
Retinoic acid (RA), a metabolite of vitamin A, has been found to influence regeneration in the adult central nervous system (CNS). There may be an effect of RA in the recovery/repair in multiple sclerosis (MS), an autoimmune and neurodegenerative disease of the CNS. We hypothesized that RA is a regulator of the further differentiation of oligodendrocyte precursor cells (OPCs) – cells key to the remyelination process in MS. We conducted studies utilizing RNA-sequencing in human embryonic stem cell (hESC)-derived neural stem cells (NSCs) and OPCs so as to understand the role of transcriptional regulators during transition from both ESCs to NSCs and NSCs to OPCs. We identified that expression of retinoic acid receptors β and γ (RARβ and RARγ ) was significantly increased following the transition from NSCs to OPCs. We also demonstrated that long term in vitro culture of hESC-derived OPC with different isoforms of RA led to the significant up-regulation of two known transcriptional inhibitors of oligodendrocyte differentiation: Hes5 following prolonged treatment with all-trans-RA, 9-cis RA and 13-cis RA; and Id4 following treatment with 13cisRA. These results suggest that long term exposure to certain RA isoforms may impact the continued differentiation of this population. Retinoic acid (ATRA) might have an effect on generation of oligodendrocyte precursor cells (OPC) in the CNS. RNA-sequencing used for identification of up-regulation of RARβ and RARɣ expression during OPC differentiation. Activation of RARβ and RARɣ by using three different agonists led increase expression of Hes5 and Id4.
Collapse
Affiliation(s)
- Sun Young Kim
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA.,Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Eve E Kelland
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Ji Hong Kim
- Department of Psychiatry and Behavioral Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.,Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Brett T Lund
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Xiao Chang
- Department of Psychiatry and Behavioral Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.,Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Kai Wang
- Department of Psychiatry and Behavioral Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.,Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.,Division of Bioinformatics, Department of Preventive medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Leslie P Weiner
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA.,Department of Molecular Microbiology & Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.,Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
27
|
Tse KH, Herrup K. DNA damage in the oligodendrocyte lineage and its role in brain aging. Mech Ageing Dev 2016; 161:37-50. [PMID: 27235538 DOI: 10.1016/j.mad.2016.05.006] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 05/23/2016] [Accepted: 05/25/2016] [Indexed: 11/25/2022]
Abstract
Myelination is a recent evolutionary addition that significantly enhances the speed of transmission in the neural network. Even slight defects in myelin integrity impair performance and enhance the risk of neurological disorders. Indeed, myelin degeneration is an early and well-recognized neuropathology that is age associated, but appears before cognitive decline. Myelin is only formed by fully differentiated oligodendrocytes, but the entire oligodendrocyte lineage are clear targets of the altered chemistry of the aging brain. As in neurons, unrepaired DNA damage accumulates in the postmitotic oligodendrocyte genome during normal aging, and indeed may be one of the upstream causes of cellular aging - a fact well illustrated by myelin co-morbidity in premature aging syndromes arising from deficits in DNA repair enzymes. The clinical and experimental evidence from Alzheimer's disease, progeroid syndromes, ataxia-telangiectasia and other conditions strongly suggest that oligodendrocytes may in fact be uniquely vulnerable to oxidative DNA damage. If this damage remains unrepaired, as is increasingly true in the aging brain, myelin gene transcription and oligodendrocyte differentiation is impaired. Delineating the relationships between early myelin loss and DNA damage in brain aging will offer an additional dimension outside the neurocentric view of neurodegenerative disease.
Collapse
Affiliation(s)
- Kai-Hei Tse
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong.
| | - Karl Herrup
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong
| |
Collapse
|
28
|
Abstract
Id (DNA binding and/or differentiation) proteins occur physiologically during ontogenesis and negatively regulate the activity of other helix-loop-helix (HLH) proteins. Id2 protein causes block of cells differentiation in the S phase of the cell cycle and regulates the activity of Rb protein. The role of Id2 protein in physiological cell cycle progression and in neuroblastoma (NBL) pathogenesis was proposed by Lasorella. The aim of the study was evaluation of Id2 expression and its prognostic significance in NBL cells coming from primary tumors and evaluation of its prognostic significance, and correlation of Id2 expression with known prognostic factors. Sixty patients with primary NBL treated from 1991 to 2005 were included in the analysis. We found 50 patients with high and 10 patients with low intensity of Id2 expression. The median percentage of NBL cells with Id2 expression was 88 %. We found no correlation between the number of NBL cells or the intensity of Id2 expression and OS and DFS. In patients with stage 4 NBL, almost all patients had high expression of Id2 and it was significantly more common than in other disease stages (p = 0,03). We found no correlation between Id2 expression and other known prognostic factor in NBL patients. We assume that Id2 is not prognostic factor. However, due to its abundant expression in most of NBL cells and its role in cell cycle, it may be potential therapeutic target. Exact knowledge of expression time may be helpful in explaining mechanisms of oncogenesis.
Collapse
Affiliation(s)
- Aleksandra Wieczorek
- Department of Pediatric Oncology and Hematology, Polish-American Institute of Pediatrics, Jagiellonian University Medical College, Krakow, Poland,
| | | |
Collapse
|
29
|
Zhao C, Ma D, Zawadzka M, Fancy SPJ, Elis-Williams L, Bouvier G, Stockley JH, de Castro GM, Wang B, Jacobs S, Casaccia P, Franklin RJM. Sox2 Sustains Recruitment of Oligodendrocyte Progenitor Cells following CNS Demyelination and Primes Them for Differentiation during Remyelination. J Neurosci 2015; 35:11482-99. [PMID: 26290228 PMCID: PMC6605237 DOI: 10.1523/jneurosci.3655-14.2015] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Revised: 05/27/2015] [Accepted: 06/21/2015] [Indexed: 12/16/2022] Open
Abstract
The Sox family of transcription factors have been widely studied in the context of oligodendrocyte development. However, comparatively little is known about the role of Sox2, especially during CNS remyelination. Here we show that the expression of Sox2 occurs in oligodendrocyte progenitor cells (OPCs) in rodent models during myelination and in activated adult OPCs responding to demyelination, and is also detected in multiple sclerosis lesions. In normal adult white matter of both mice and rats, it is neither expressed by adult OPCs nor by oligodendrocytes (although it is expressed by a subpopulation of adult astrocytes). Overexpression of Sox2 in rat OPCs in vitro maintains the cells in a proliferative state and inhibits differentiation, while Sox2 knockout results in decreased OPC proliferation and survival, suggesting that Sox2 contributes to the expansion of OPCs during the recruitment phase of remyelination. Loss of function in cultured mouse OPCs also results in an impaired ability to undergo normal differentiation in response to differentiation signals, suggesting that Sox2 expression in activated OPCs also primes these cells to eventually undergo differentiation. In vivo studies on remyelination following experimental toxin-induced demyelination in mice with inducible loss of Sox2 revealed impaired remyelination, which was largely due to a profound attenuation of OPC recruitment and likely also due to impaired differentiation. Our results reveal a key role of Sox2 expression in OPCs responding to demyelination, enabling them to effectively contribute to remyelination. SIGNIFICANCE STATEMENT Understanding the mechanisms of CNS remyelination is central to developing effective means by which this process can be therapeutically enhanced in chronic demyelinating diseases such as multiple sclerosis. In this study, we describe the role of Sox2, a transcription factor widely implicated in stem cell biology, in CNS myelination and remyelination. We show how Sox2 is expressed in oligodendrocyte progenitor cells (OPCs) preparing to undergo differentiation, allowing them to undergo proliferation and priming them for subsequent differentiation. Although Sox2 is unlikely to be a direct therapeutic target, these data nevertheless provide more information on how OPC differentiation is controlled and therefore enriches our understanding of this important CNS regenerative process.
Collapse
Affiliation(s)
- Chao Zhao
- Wellcome Trust-Medical Research Council Stem Cell Institute and Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0AH, United Kingdom, and
| | - Dan Ma
- Wellcome Trust-Medical Research Council Stem Cell Institute and Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0AH, United Kingdom, and
| | - Malgorzata Zawadzka
- Wellcome Trust-Medical Research Council Stem Cell Institute and Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0AH, United Kingdom, and
| | - Stephen P J Fancy
- Wellcome Trust-Medical Research Council Stem Cell Institute and Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0AH, United Kingdom, and
| | - Lowri Elis-Williams
- Wellcome Trust-Medical Research Council Stem Cell Institute and Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0AH, United Kingdom, and
| | - Guy Bouvier
- Wellcome Trust-Medical Research Council Stem Cell Institute and Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0AH, United Kingdom, and
| | - John H Stockley
- Wellcome Trust-Medical Research Council Stem Cell Institute and Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0AH, United Kingdom, and
| | - Glaucia Monteiro de Castro
- Wellcome Trust-Medical Research Council Stem Cell Institute and Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0AH, United Kingdom, and
| | - Bowei Wang
- Wellcome Trust-Medical Research Council Stem Cell Institute and Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0AH, United Kingdom, and
| | - Sabrina Jacobs
- Wellcome Trust-Medical Research Council Stem Cell Institute and Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0AH, United Kingdom, and
| | - Patrizia Casaccia
- Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029-6574
| | - Robin J M Franklin
- Wellcome Trust-Medical Research Council Stem Cell Institute and Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0AH, United Kingdom, and
| |
Collapse
|
30
|
PAK5-mediated E47 phosphorylation promotes epithelial-mesenchymal transition and metastasis of colon cancer. Oncogene 2015. [PMID: 26212009 DOI: 10.1038/onc.2015.259] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The p21-activated kinase 5 (PAK5) is overexpressed in advanced cancer and the transcription factor E47 is a direct repressor of E-cadherin and inducer of epithelial-mesenchymal transition (EMT). However, the relationship between PAK5 and E47 has not been explored. In this study, we found that PAK5-mediated E47 phosphorylation promoted EMT in advanced colon cancer. PAK5 interacted with E47 and phosphorylated E47 on Ser39 under hepatocyte growth factor (HGF) stimulation, which decreased cell-cell cohesion, increased cell migration and invasion in vitro and promoted metastasis in a xenograft model. Furthermore, phosphorylation of E47 facilitated its accumulating in nucleus in an importin α-dependent manner, and enhanced E47 binding to E-cadherin promoter directly, leading to inhibition of E-cadherin transcription. In contrast, PAK5-knockdown resulted in blockage of HGF-induced E47 phosphorylation, attenuated association of E47 with importin α and decreased E47 binding to E-cadherin promoter. In addition, we demonstrated a close correlation between PAK5 and phospho-Ser39 E47 expression in colon cancer specimens. More importantly, high expression of phospho-E47 was associated with an aggressive phenotype of colon cancer and nuclear phospho-E47 staining was found in certain cases of colon cancer with metastasis. Collectively, E47 is a novel substrate of PAK5, and PAK5-mediated phosphorylation of E47 promotes EMT and metastasis of colon cancer, suggesting that phosphorylated E47 on Ser39 may be a potential therapeutic target in progressive colon cancer.
Collapse
|
31
|
Takebayashi H, Ikenaka K. Oligodendrocyte generation during mouse development. Glia 2015; 63:1350-6. [PMID: 26013243 DOI: 10.1002/glia.22863] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 04/30/2015] [Indexed: 12/15/2022]
Abstract
Oligodendrocytes (OLs) are glial cells, which generate myelin in the central nervous system. Their interesting developmental features attract many neurobiologists eager to study cell differentiation, gene expression regulation, or dynamic morphogenesis. Their primary role in protecting the axons has major impacts in the medical research field: in multiple sclerosis, a demyelinating disease in which remyelination is blocked. Oligodendrogenesis is involved in higher brain function including motor skill learning and cognitive function. Here, we review advances in the research on OL development and highlight areas where questions remain to be answered in both developmental biology and neurobiology related aspects.
Collapse
Affiliation(s)
| | - Kazuhiro Ikenaka
- Division of Neurobiology and Bioinformatics, National Institute for Physiological Sciences, Okazaki, Japan
| |
Collapse
|
32
|
Ferrer-Vicens I, Riffo‐Campos ÁL, Zaragozá R, García C, López-Rodas G, Viña JR, Torres L, García-Trevijano ER. In vivo genome-wide binding of Id2 to E2F4 target genes as part of a reversible program in mice liver. Cell Mol Life Sci 2014; 71:3583-97. [PMID: 24573694 PMCID: PMC11113549 DOI: 10.1007/s00018-014-1588-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Revised: 01/30/2014] [Accepted: 02/12/2014] [Indexed: 12/14/2022]
Abstract
The inhibitor of differentiation Id2, a protein lacking the basic DNA-binding domain, is involved in the modulation of a number of biological processes. The molecular mechanisms explaining Id2 pleiotropic functions are poorly understood. Id2 and E2F4 are known to bind simultaneously to c-myc promoter. To study whether Id2 plays a global role on transcriptional regulation, we performed in vivo genome-wide ChIP/chip experiments for Id2 and E2F4 in adult mouse liver. An Id2-containing complex was bound to a common sequence downstream from the TSS on a subset of 442 E2F4 target genes mainly related to cell development and chromatin structure. We found a positive correlation between Id2 protein levels and the expression of E2F4/Id2 targets in fetal and adult liver. Id2 protein stability increased in fetal liver by interaction with USP1 de-ubiquitinating enzyme, which was induced during development. In adult liver, USP1 and Id2 levels dramatically decreased. In differentiated liver tissue, when Id2 concentration was low, E2F4/Id2 was bound to the same region as paused Pol II and target genes remained transcriptionally inactive. Conversely, in fetal liver when Id2 levels were increased, Id2 and Pol II were released from gene promoters and target genes up-regulated. During liver regeneration after partial hepatectomy, we obtained the same results as in fetal liver. Our results suggest that Id2 might be part of a reversible development-related program involved in the paused-ON/OFF state of Pol II on selected genes that would remain responsive to specific stimuli.
Collapse
Affiliation(s)
- Ivan Ferrer-Vicens
- Departamento de Bioquímica y Biología Molecular. Facultad de Medicina. Fundación Investigación Hospital Clínico-INCLIVA, Universidad de Valencia, Avda. Blasco Ibañez, 17, 46010 Valencia, Spain
| | - Ángela L. Riffo‐Campos
- Facultad de Ciencias Biológicas. Fundación Investigación Hospital Clínico-INCLIVA, Universidad de Valencia, Avda. Blasco Ibañez, 17, 46010 Valencia, Spain
| | - Rosa Zaragozá
- Departamento de Bioquímica y Biología Molecular. Facultad de Medicina. Fundación Investigación Hospital Clínico-INCLIVA, Universidad de Valencia, Avda. Blasco Ibañez, 17, 46010 Valencia, Spain
| | - Concha García
- Departamento de Bioquímica y Biología Molecular. Facultad de Medicina. Fundación Investigación Hospital Clínico-INCLIVA, Universidad de Valencia, Avda. Blasco Ibañez, 17, 46010 Valencia, Spain
| | - Gerardo López-Rodas
- Facultad de Ciencias Biológicas. Fundación Investigación Hospital Clínico-INCLIVA, Universidad de Valencia, Avda. Blasco Ibañez, 17, 46010 Valencia, Spain
| | - Juan R. Viña
- Departamento de Bioquímica y Biología Molecular. Facultad de Medicina. Fundación Investigación Hospital Clínico-INCLIVA, Universidad de Valencia, Avda. Blasco Ibañez, 17, 46010 Valencia, Spain
| | - Luis Torres
- Departamento de Bioquímica y Biología Molecular. Facultad de Medicina. Fundación Investigación Hospital Clínico-INCLIVA, Universidad de Valencia, Avda. Blasco Ibañez, 17, 46010 Valencia, Spain
| | - Elena R. García-Trevijano
- Departamento de Bioquímica y Biología Molecular. Facultad de Medicina. Fundación Investigación Hospital Clínico-INCLIVA, Universidad de Valencia, Avda. Blasco Ibañez, 17, 46010 Valencia, Spain
| |
Collapse
|
33
|
Nonneman A, Robberecht W, Den Bosch LV. The role of oligodendroglial dysfunction in amyotrophic lateral sclerosis. Neurodegener Dis Manag 2014; 4:223-39. [DOI: 10.2217/nmt.14.21] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
SUMMARY Healthy oligodendrocytes are crucial for neurons and abnormal functioning of these cells is involved in several neurodegenerative diseases. We will focus on oligodendroglial pathology in amyotrophic lateral sclerosis (ALS), an adult-onset progressive neurodegenerative disease characterized by selective motor neuron loss. Recent discoveries shed new light on the crucial role of oligodendrocytes in this fatal disease. We will first give an overview of the importance of good-functioning oligodendrocytes for neuronal health, in particular for motor neurons. Subsequently, we will discuss the recent data on oligodendroglial abnormalities in ALS. We conclude that oligodendrocytes should be considered as important contributors to motor neuron degeneration. As a consequence, oligodendrocytes are a promising new therapeutic target for ALS and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Annelies Nonneman
- KU Leuven – University of Leuven, Department of Neurosciences, Experimental Neurology and Leuven Research Institute for Neuroscience and Disease (LIND), B-3000 Leuven, Belgium
- VIB, Vesalius Research Center, Laboratory of Neurobiology, B-3000 Leuven, Belgium
| | - Wim Robberecht
- KU Leuven – University of Leuven, Department of Neurosciences, Experimental Neurology and Leuven Research Institute for Neuroscience and Disease (LIND), B-3000 Leuven, Belgium
- VIB, Vesalius Research Center, Laboratory of Neurobiology, B-3000 Leuven, Belgium
- University Hospitals Leuven, Department of Neurology, B-3000 Leuven, Belgium
| | - Ludo Van Den Bosch
- KU Leuven – University of Leuven, Department of Neurosciences, Experimental Neurology and Leuven Research Institute for Neuroscience and Disease (LIND), B-3000 Leuven, Belgium
- VIB, Vesalius Research Center, Laboratory of Neurobiology, B-3000 Leuven, Belgium
| |
Collapse
|
34
|
Genomic analysis of diffuse intrinsic pontine gliomas identifies three molecular subgroups and recurrent activating ACVR1 mutations. Nat Genet 2014; 46:451-6. [PMID: 24705254 PMCID: PMC3997489 DOI: 10.1038/ng.2936] [Citation(s) in RCA: 475] [Impact Index Per Article: 47.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Accepted: 03/05/2014] [Indexed: 12/19/2022]
Abstract
Diffuse Intrinsic Pontine Glioma (DIPG) is a fatal brain cancer that arises in the brainstem of children with no effective treatment and near 100% fatality. The failure of most therapies can be attributed to the delicate location of these tumors and choosing therapies based on assumptions that DIPGs are molecularly similar to adult disease. Recent studies have unraveled the unique genetic make-up of this brain cancer with nearly 80% harboring a K27M-H3.3 or K27M-H3.1 mutation. However, DIPGs are still thought of as one disease with limited understanding of the genetic drivers of these tumors. To understand what drives DIPGs we integrated whole-genome-sequencing with methylation, expression and copy-number profiling, discovering that DIPGs are three molecularly distinct subgroups (H3-K27M, Silent, MYCN) and uncovering a novel recurrent activating mutation in the activin receptor ACVR1, in 20% of DIPGs. Mutations in ACVR1 were constitutively activating, leading to SMAD phosphorylation and increased expression of downstream activin signaling targets ID1 and ID2. Our results highlight distinct molecular subgroups and novel therapeutic targets for this incurable pediatric cancer.
Collapse
|
35
|
Qu X, Qi D, Dong F, Wang B, Guo R, Luo M, Yao R. Quercetin improves hypoxia-ischemia induced cognitive deficits via promoting remyelination in neonatal rat. Brain Res 2014; 1553:31-40. [PMID: 24480472 DOI: 10.1016/j.brainres.2014.01.035] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Revised: 01/07/2014] [Accepted: 01/20/2014] [Indexed: 12/26/2022]
Abstract
Myelination failure is associated with perinatal cerebral hypoxia-ischemia (PHI) induced brain injury in premature infants. How to efficiently promote remyelination is crucial for improving cognitive deficits caused by brain injury. Here, we demonstrated that quercetin (Que), a kind of flavonoids, significantly improved cognitive deficits and the behavior of PHI-rat in Morris water maze and open field tasks. After administration of Que to PHI-rat, the number of neogenetic Olig2⁺ oligodendrocyte progenitor cells (OPCs) was evidently increased in the subventricular zone. Additionally, in corpus callosum (CC), the expression of MBP (myelin basic protein) was increased, and the myelin sheaths reached normal level at 30 days with more compact while less damaged myelin sheaths and more mature oligodendrocytes (OLs) repopulating the CC compared with PHI groups. In a word, our findings indicated that Que could remarkably improve both cognition performance and myelination in the context of PHI-induced brain injury by promoting the proliferation of OPCs and strengthening survival of OLs in vivo.
Collapse
Affiliation(s)
- Xuebin Qu
- Department of Neurobiology, Xuzhou Medical College, Xuzhou 221009, Jiangsu, PR China
| | - Dashi Qi
- Department of Neurobiology, Xuzhou Medical College, Xuzhou 221009, Jiangsu, PR China
| | - Fuxing Dong
- Department of Neurobiology, Xuzhou Medical College, Xuzhou 221009, Jiangsu, PR China
| | - Bei Wang
- Department of Neurobiology, Xuzhou Medical College, Xuzhou 221009, Jiangsu, PR China
| | - Rui Guo
- Department of Neurobiology, Xuzhou Medical College, Xuzhou 221009, Jiangsu, PR China
| | - Mengjiao Luo
- Department of Neurobiology, Xuzhou Medical College, Xuzhou 221009, Jiangsu, PR China
| | - Ruiqin Yao
- Department of Neurobiology, Xuzhou Medical College, Xuzhou 221009, Jiangsu, PR China.
| |
Collapse
|
36
|
Promoting return of function in multiple sclerosis: An integrated approach. Mult Scler Relat Disord 2013; 2:S2211-0348(13)00044-8. [PMID: 24363985 DOI: 10.1016/j.msard.2013.04.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Multiple sclerosis is a disease characterized by inflammatory demyelination, axonal degeneration and progressive brain atrophy. Most of the currently available disease modifying agents proved to be very effective in managing the relapse rate, however progressive neuronal damage continues to occur and leads to progressive accumulation of irreversible disability. For this reason, any therapeutic strategy aimed at restoration of function must take into account not only immunomodulation, but also axonal protection and new myelin formation. We further highlight the importance of an holistic approach, which considers the variability of therapeutic responsiveness as the result of the interplay between genetic differences and the epigenome, which is in turn affected by gender, age and differences in life style including diet, exercise, smoking and social interaction.
Collapse
|
37
|
Plemel JR, Manesh SB, Sparling JS, Tetzlaff W. Myelin inhibits oligodendroglial maturation and regulates oligodendrocytic transcription factor expression. Glia 2013; 61:1471-87. [PMID: 23839973 DOI: 10.1002/glia.22535] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2012] [Revised: 04/20/2013] [Accepted: 04/26/2013] [Indexed: 12/12/2022]
Abstract
Myelin loss is a hallmark of multiple sclerosis (MS) and promoting central nervous system myelin repair has become a major therapeutic target. Despite the presence of oligodendrocytes precursors cells (OPCs) in chronic lesions of MS, remyelination often fails. The mechanism underlying this failure of remyelination remains unknown, but it is hypothesized that environmental cues act to inhibit the maturation/differentiation of oligodendroglia, preventing remyelination. The rate of CNS remyelination is correlated to the speed of phagocytosis of myelin debris, which is present following demyelination and trauma. Thus, myelin debris could inhibit CNS remyelination. Here, we demonstrate that OPCs cultured on myelin were robustly inhibited in their maturation, as characterized by the decreased expression of immature and mature oligodendrocytes markers, the impaired production of myelin gene products, as well as their stalled morphological complexity relative to OPCs cultured on a control substrate. OPCs in contact with myelin stopped proliferating and decreased the expression of OPC markers to a comparable degree as cells grown on a control substrate. The expression of two transcription factors known to prevent OPC differentiation and maturation were increased in cells that were in contact with myelin: inhibitor of differentiation family (ID) members 2 and 4. Overexpression of ID2 and ID4 in OPCs was previously reported to decrease the percentage of cells expressing mature oligodendrocyte markers. However, knockdown of ID2 and/or ID4 in OPCs did not increase oligodendroglial maturation on or off of myelin, suggesting that contact with myelin regulates additional regulatory elements.
Collapse
Affiliation(s)
- Jason R Plemel
- ICORD (International Collaboration on Repair Discoveries), Blusson Spinal Cord Centre, Vancouver, British Columbia, Canada
| | | | | | | |
Collapse
|
38
|
Improving myelin/oligodendrocyte-related dysfunction: a new mechanism of antipsychotics in the treatment of schizophrenia? Int J Neuropsychopharmacol 2013; 16:691-700. [PMID: 23164411 DOI: 10.1017/s1461145712001095] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Schizophrenia is a severe psychiatric disorder with complex clinical manifestations and its aetiological factors remain unclear. During the past decade, the oligodendrocyte-related myelin dysfunction was proposed as a hypothesis for schizophrenia, supported initially by a series of neuroimaging studies and genetic evidence. Recently, the effects of antipsychotics on myelination and oligodendroglial lineage development and their underlying molecular mechanisms were evaluated. Data from those studies suggest that the antipsychotics-resulting improvement in myelin/oligodendrocyte-related dysfunction may contribute, at least in part, to their therapeutic effect on schizophrenia. Importantly, these findings may provide the basis for a new insight into the therapeutic strategy by targeting the oligodendroglia lineage cells against schizophrenia.
Collapse
|
39
|
Zuo H, Nishiyama A. Polydendrocytes in development and myelin repair. Neurosci Bull 2013; 29:165-76. [PMID: 23516142 DOI: 10.1007/s12264-013-1320-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2012] [Accepted: 01/30/2013] [Indexed: 11/30/2022] Open
Abstract
Polydendrocytes (NG2 cells) are a distinct type of glia that populate the developing and adult central nervous systems (CNS). In the adult CNS, they retain mitotic activity and represent the largest proliferating cell population. Genetic and epigenetic mechanisms regulate the fate of polydendrocytes, which give rise to both oligodendrocytes and astrocytes. In addition, polydendrocytes actively differentiate into myelin-forming oligodendrocytes in response to demyelination. This review summarizes the current knowledge regarding polydendrocyte development, which provides an important basis for understanding the mechanisms that lead to the remyelination of demyelinated lesions.
Collapse
Affiliation(s)
- Hao Zuo
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT 06269-3156, USA
| | | |
Collapse
|
40
|
mTOR: a link from the extracellular milieu to transcriptional regulation of oligodendrocyte development. ASN Neuro 2013; 5:e00108. [PMID: 23421405 PMCID: PMC3601842 DOI: 10.1042/an20120092] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Oligodendrocyte development is controlled by numerous extracellular signals that regulate a series of transcription factors that promote the differentiation of oligodendrocyte progenitor cells to myelinating cells in the central nervous system. A major element of this regulatory system that has only recently been studied is the intracellular signalling from surface receptors to transcription factors to down-regulate inhibitors and up-regulate inducers of oligodendrocyte differentiation and myelination. The current review focuses on one such pathway: the mTOR (mammalian target of rapamycin) pathway, which integrates signals in many cell systems and induces cell responses including cell proliferation and cell differentiation. This review describes the known functions of mTOR as they relate to oligodendrocyte development, and its recently discovered impact on oligodendrocyte differentiation and myelination. A potential model for its role in oligodendrocyte development is proposed.
Collapse
|
41
|
Havrda MC, Paolella BR, Ward NM, Holroyd KB. Behavioral abnormalities and Parkinson's-like histological changes resulting from Id2 inactivation in mice. Dis Model Mech 2012; 6:819-27. [PMID: 23264561 PMCID: PMC3634664 DOI: 10.1242/dmm.010041] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Characterizing dopaminergic neuronal development and function in novel genetic animal models might uncover strategies for researchers to develop disease-modifying treatments for neurologic disorders. Id2 is a transcription factor expressed in the developing central nervous system. Id2(-/-) mice have fewer dopaminergic neurons in the olfactory bulb and reduced olfactory discrimination, a pre-clinical marker of Parkinson's disease. Here, we summarize behavioral, histological and in vitro molecular biological analyses to determine whether midbrain dopaminergic neurons are affected by Id2 loss. Id2(-/-) mice were hyperactive at 1 and 3 months of age, but by 6 months showed reduced activity. Id2(-/-) mice showed age-dependent histological alterations in dopaminergic neurons of the substantia nigra pars compacta (SNpC) associated with changes in locomotor activity. Reduced dopamine transporter (DAT) expression was observed at early ages in Id2(-/-) mice and DAT expression was dependent on Id2 expression in an in vitro dopaminergic differentiation model. Evidence of neurodegeneration, including activated caspase-3 and glial infiltration, were noted in the SNpC of older Id2(-/-) mice. These findings document a novel role for Id2 in the maintenance of midbrain dopamine neurons. The Id2(-/-) mouse should provide unique opportunities to study the progression of neurodegenerative disorders involving the dopamine system.
Collapse
Affiliation(s)
- Matthew C Havrda
- Norris Cotton Cancer Center and Department of Pediatrics, Geisel School of Medicine at Dartmouth, Dartmouth-Hitchcock Medical Center, Lebanon, NH 03756, USA.
| | | | | | | |
Collapse
|