1
|
Hai Q, Wang Y, Li H, Pei H, Wang N, Zhang X, Fan M, Liao J, Wen W, Zhao J, Yang L, Cui H. Plantamajoside Promotes NGF/TrkA Pathway to Inhibit Neuronal Apoptosis and Improve Diabetic Peripheral Neuropathy. J Cell Mol Med 2025; 29:e70571. [PMID: 40289624 PMCID: PMC12034938 DOI: 10.1111/jcmm.70571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 04/15/2025] [Accepted: 04/17/2025] [Indexed: 04/30/2025] Open
Abstract
The symptoms caused by diabetic peripheral neuropathy (DPN) have severely impacted patients' quality of life. While plantamajoside (PMS) exhibits neuroprotective properties, its efficacy and molecular mechanisms against DPN are unexplored. This study first established a high glucose (HG)-induced in vitro model of DPN and investigated the neuroprotective effects of PMS on RSC96 cells. We next demonstrated the anti-apoptotic effects of PMS and NGF/TrkA pathway mediated neurotrophic effects. Finally, we established a DPN mouse model and confirmed the therapeutic effects of PMS on DPN mice through behavioural tests and pathological staining, while also assessing the impact of PMS on the NGF/TrkA pathway and apoptosis. Our results showed that, in HG-induced DPN models, PMS enhanced cell viability while reducing LDH activity. Transcriptomics results indicated that the Apoptosis and Neurotrophins signalling pathways were key pathways for PMS on DPN. PMS treatment reduced HG-induced RSC96 cell apoptosis while enhancing NGF levels and upregulating NGF/TrkA-related protein expression. However, this protection was abolished by TrkA inhibitor or NGF neutralising antibodies. In vivo experimental results showed that PMS improved the mechanical pain threshold, thermal pain reaction time, and nerve conduction velocity of DPN mice. PMS improved pathological damage to the sciatic nerve, enhanced the number of Nissl bodies, reduced TUNEL-positive expression, and upregulated NGF levels. Furthermore, PMS reduced apoptosis and elevated NGF/TrkA-related protein expression in the sciatic nerve of DPN mice. In conclusion, PMS alleviates DPN through activating the NGF/TrkA pathway and inhibiting apoptosis.
Collapse
Affiliation(s)
- Qingshan Hai
- Nanjing University of Chinese MedicineNanjinChina
- Basic Medical SchoolYunnan University of Chinese MedicineKunmingChina
| | - Yuming Wang
- Tianjin University of Traditional Chinese MedicineTianjinChina
| | - Hanzhou Li
- Tianjin University of Traditional Chinese MedicineTianjinChina
| | - Huan Pei
- Basic Medical SchoolYunnan University of Chinese MedicineKunmingChina
| | - Ning Wang
- First School of Clinical MedicineYunnan University of Chinese MedicineKunmingChina
| | - Xiaoxia Zhang
- Basic Medical SchoolYunnan University of Chinese MedicineKunmingChina
| | - Mingyao Fan
- Basic Medical SchoolYunnan University of Chinese MedicineKunmingChina
| | - Jiabao Liao
- First School of Clinical MedicineYunnan University of Chinese MedicineKunmingChina
| | - Weibo Wen
- Nanjing University of Chinese MedicineNanjinChina
- First School of Clinical MedicineYunnan University of Chinese MedicineKunmingChina
| | - Jie Zhao
- First School of Clinical MedicineYunnan University of Chinese MedicineKunmingChina
| | - Ling Yang
- School of NursingYunnan University of Chinese MedicineKunmingChina
| | - Huantian Cui
- First School of Clinical MedicineYunnan University of Chinese MedicineKunmingChina
| |
Collapse
|
2
|
Huber M, Brummer T. Enzyme Is the Name-Adapter Is the Game. Cells 2024; 13:1249. [PMID: 39120280 PMCID: PMC11311582 DOI: 10.3390/cells13151249] [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: 04/30/2024] [Revised: 07/15/2024] [Accepted: 07/16/2024] [Indexed: 08/10/2024] Open
Abstract
Signaling proteins in eukaryotes usually comprise a catalytic domain coupled to one or several interaction domains, such as SH2 and SH3 domains. An additional class of proteins critically involved in cellular communication are adapter or scaffold proteins, which fulfill their purely non-enzymatic functions by organizing protein-protein interactions. Intriguingly, certain signaling enzymes, e.g., kinases and phosphatases, have been demonstrated to promote particular cellular functions by means of their interaction domains only. In this review, we will refer to such a function as "the adapter function of an enzyme". Though many stories can be told, we will concentrate on several proteins executing critical adapter functions in cells of the immune system, such as Bruton´s tyrosine kinase (BTK), phosphatidylinositol 3-kinase (PI3K), and SH2-containing inositol phosphatase 1 (SHIP1), as well as in cancer cells, such as proteins of the rat sarcoma/extracellular signal-regulated kinase (RAS/ERK) mitogen-activated protein kinase (MAPK) pathway. We will also discuss how these adaptor functions of enzymes determine or even undermine the efficacy of targeted therapy compounds, such as ATP-competitive kinase inhibitors. Thereby, we are highlighting the need to develop pharmacological approaches, such as proteolysis-targeting chimeras (PROTACs), that eliminate the entire protein, and thus both enzymatic and adapter functions of the signaling protein. We also review how genetic knock-out and knock-in approaches can be leveraged to identify adaptor functions of signaling proteins.
Collapse
Affiliation(s)
- Michael Huber
- Institute of Biochemistry and Molecular Immunology, Medical Faculty, RWTH Aachen University, 52074 Aachen, Germany
| | - Tilman Brummer
- Institute of Molecular Medicine and Cell Research, IMMZ, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Center for Biological Signalling Studies BIOSS, University of Freiburg, 79104 Freiburg, Germany
| |
Collapse
|
3
|
Emery B, Wood TL. Regulators of Oligodendrocyte Differentiation. Cold Spring Harb Perspect Biol 2024; 16:a041358. [PMID: 38503504 PMCID: PMC11146316 DOI: 10.1101/cshperspect.a041358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2024]
Abstract
Myelination has evolved as a mechanism to ensure fast and efficient propagation of nerve impulses along axons. Within the central nervous system (CNS), myelination is carried out by highly specialized glial cells, oligodendrocytes. The formation of myelin is a prolonged aspect of CNS development that occurs well into adulthood in humans, continuing throughout life in response to injury or as a component of neuroplasticity. The timing of myelination is tightly tied to the generation of oligodendrocytes through the differentiation of their committed progenitors, oligodendrocyte precursor cells (OPCs), which reside throughout the developing and adult CNS. In this article, we summarize our current understanding of some of the signals and pathways that regulate the differentiation of OPCs, and thus the myelination of CNS axons.
Collapse
Affiliation(s)
- Ben Emery
- Jungers Center for Neurosciences Research, Department of Neurology, Oregon Health & Science University, Portland, Oregon 97239, USA
| | - Teresa L Wood
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers University, Newark, New Jersey 07103, USA
| |
Collapse
|
4
|
Srivastava K, Mishra R. Pax6 affects Ras-Raf-ERK1/2 in mouse aging brain. Biogerontology 2023; 24:901-912. [PMID: 37436500 DOI: 10.1007/s10522-023-10044-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 06/06/2023] [Indexed: 07/13/2023]
Abstract
Pax6, a transcription factor and multifunctional protein, changes during aging. It also interacts with regulator proteins involved in cell metabolism and survival signalling pathways including Ras-GAP. Many forms of Ras, Raf and ERK1/2 are known but information on their region-specific expression patterns are unavailable from brain during aging. Therefore, it has been intended to evaluate expressions of Pax6 and forms of Ras, Raf, ERK1/2 in hippocampus, caudate nucleus, amygdale, cerebral cortex, cerebellum and olfactory lobe. Association of Pax6 with Ras, Raf and ERK1/2 was evaluated in co-culture (PC-12, C6-glia, U-87 MG) of neuroglia cell lines. Impacts of Pax6 were evaluated by siRNA mediated knockdown and expression patterns Ras-Raf-Erk1/2. Analysis of activities of Pax6 and impacts of 5'AMP, wild-type and mutant ERK were done by RT-PCR and luciferase reporter assay. Results indicate age-dependent changes of Pax6, Ras, Raf, ERK1/2 in different regions of brain of young and old mice. Erk1/2 shows synergistic activities to Pax6.
Collapse
Affiliation(s)
- Khushboo Srivastava
- Biochemistry and Molecular Biology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Rajnikant Mishra
- Biochemistry and Molecular Biology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, India.
| |
Collapse
|
5
|
Ye Q, Srivastava P, Al-Kuwari N, Chen X. Oncogenic BRAFV600E induces microglial proliferation through extracellular signal-regulated kinase and neuronal death through c-Jun N-terminal kinase. Neural Regen Res 2023; 18:1613-1622. [PMID: 36571370 PMCID: PMC10075110 DOI: 10.4103/1673-5374.361516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 06/13/2022] [Accepted: 10/18/2022] [Indexed: 11/19/2022] Open
Abstract
Activating V600E in v-Raf murine sarcoma viral oncogene homolog B (BRAF) is a common driver mutation in cancers of multiple tissue origins, including melanoma and glioma. BRAFV600E has also been implicated in neurodegeneration. The present study aims to characterize BRAFV600E during cell death and proliferation of three major cell types of the central nervous system: neurons, astrocytes, and microglia. Multiple primary cultures (primary cortical mixed culture) and cell lines of glial cells (BV2) and neurons (SH-SY5Y) were employed. BRAFV600E and BRAFWT expression was mediated by lentivirus or retrovirus. Blockage of downstream effectors (extracellular signal-regulated kinase 1/2 and JNK1/2) were achieved by siRNA. In astrocytes and microglia, BRAFV600E induces cell proliferation, and the proliferative effect in microglia is mediated by activated extracellular signal-regulated kinase, but not c-Jun N-terminal kinase. Conditioned medium from BRAFV600E-expressing microglia induced neuronal death. In neuronal cells, BRAFV600E directly induces neuronal death, through c-Jun N-terminal kinase but not extracellular signal-regulated kinase. We further show that BRAF-related genes are enriched in pathways in patients with Parkinson's disease. Our study identifies distinct consequences mediated by distinct downstream effectors in dividing glial cells and in neurons following the same BRAF mutational activation and a causal link between BRAF-activated microglia and neuronal cell death that does not require physical proximity. It provides insight into a possibly important role of BRAF in neurodegeneration as a result of either dysregulated BRAF in neurons or its impact on glial cells.
Collapse
Affiliation(s)
- Qing Ye
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
- Department of Neurology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Pranay Srivastava
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Nasser Al-Kuwari
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Xiqun Chen
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| |
Collapse
|
6
|
CRISPR/CasRx-Mediated RNA Knockdown Reveals That ACE2 Is Involved in the Regulation of Oligodendroglial Cell Morphological Differentiation. Noncoding RNA 2022; 8:ncrna8030042. [PMID: 35736639 PMCID: PMC9229887 DOI: 10.3390/ncrna8030042] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 05/27/2022] [Accepted: 06/03/2022] [Indexed: 12/05/2022] Open
Abstract
Angiotensin-converting enzyme 2 (ACE2) plays a role in catalyzing angiotensin II conversion to angiotensin (1–7), which often counteracts the renin-angiotensin system. ACE2 is expressed not only in the cells of peripheral tissues such as the heart and kidney, but also in those of the central nervous system (CNS). Additionally, ACE2 acts as the receptor required for the entry of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), whose binding leads to endocytotic recycling and possible degradation of the ACE2 proteins themselves. One of the target cells for SARS-CoV-2 in the CNS is oligodendrocytes (oligodendroglial cells), which wrap neuronal axons with their differentiated plasma membranes called myelin membranes. Here, for the first time, we describe the role of ACE2 in FBD-102b cells, which are used as the differentiation models of oligodendroglial cells. Unexpectedly, RNA knockdown of ACE2 with CasRx-mediated gRNA or the cognate siRNA promoted oligodendroglial cell morphological differentiation with increased expression or phosphorylation levels of differentiation and/or myelin marker proteins, suggesting the negative role of ACE2 in morphological differentiation. Notably, ACE2′s intracellular region preferentially interacted with the active GTP-bound form of Ras. Thus, knockdown of ACE2 relatively increased GTP-bound Ras in an affinity-precipitation assay. Indeed, inhibition of Ras resulted in decreasing both morphological differentiation and expression or phosphorylation levels of marker proteins, confirming the positive role of Ras in differentiation. These results indicate the role of ACE2 itself as a negative regulator of oligodendroglial cell morphological differentiation, newly adding ACE2 to the list of regulators of oligodendroglial morphogenesis as well as of Ras-binding proteins. These findings might help us to understand why SARS-CoV-2 causes pathological effects in the CNS.
Collapse
|
7
|
MacLean M, López-Díez R, Vasquez C, Gugger PF, Schmidt AM. Neuronal-glial communication perturbations in murine SOD1 G93A spinal cord. Commun Biol 2022; 5:177. [PMID: 35228715 PMCID: PMC8885678 DOI: 10.1038/s42003-022-03128-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 02/08/2022] [Indexed: 12/13/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is an incurable disease characterized by proteinaceous aggregate accumulation and neuroinflammation culminating in rapidly progressive lower and upper motor neuron death. To interrogate cell-intrinsic and inter-cell type perturbations in ALS, single-nucleus RNA sequencing was performed on the lumbar spinal cord in the murine ALS model SOD1G93A transgenic and littermate control mice at peri-symptomatic onset stage of disease, age 90 days. This work uncovered perturbed tripartite synapse functions, complement activation and metabolic stress in the affected spinal cord; processes evidenced by cell death and proteolytic stress-associated gene sets. Concomitantly, these pro-damage events in the spinal cord co-existed with dysregulated reparative mechanisms. This work provides a resource of cell-specific niches in the ALS spinal cord and asserts that interwoven dysfunctional neuronal-glial communications mediating neurodegeneration are underway prior to overt disease manifestation and are recapitulated, in part, in the human post-mortem ALS spinal cord. In this paper, single-nucleus RNA sequencing was performed to provide a resource of cell-specific niches in the murine ALS model spinal cord at peri-symptomatic onset stage of disease. The data suggest that dysfunctional neuronal-glial communication occurs prior to disease onset, which is partially recapitulated in human post-mortem ALS spinal cord tissue.
Collapse
Affiliation(s)
- Michael MacLean
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Raquel López-Díez
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Carolina Vasquez
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Paul F Gugger
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Ann Marie Schmidt
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, New York, NY, 10016, USA.
| |
Collapse
|
8
|
Binamé F, Pham-Van LD, Bagnard D. Manipulating oligodendrocyte intrinsic regeneration mechanism to promote remyelination. Cell Mol Life Sci 2021; 78:5257-5273. [PMID: 34019104 PMCID: PMC11073109 DOI: 10.1007/s00018-021-03852-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 04/14/2021] [Accepted: 05/08/2021] [Indexed: 02/06/2023]
Abstract
In demyelinated lesions, astrocytes, activated microglia and infiltrating macrophages secrete several factors regulating oligodendrocyte precursor cells' behaviour. What appears to be the initiation of an intrinsic mechanism of myelin repair is only leading to partial recovery and inefficient remyelination, a process worsening over the course of the disease. This failure is largely due to the concomitant accumulation of inhibitory cues in and around the lesion sites opposing to growth promoting factors. Here starts a complex game of interactions between the signalling pathways controlling oligodendrocytes migration or differentiation. Receptors of positive or negative cues are modulating Ras, PI3K or RhoGTPases pathways acting on oligodendrocyte cytoskeleton remodelling. From the description of this intricate signalling network, this review addresses the extent to which the modulation of the global response to inhibitory cues may pave the route towards novel therapeutic approaches for myelin repair.
Collapse
Affiliation(s)
- Fabien Binamé
- INSERM U1119, Biopathology of Myelin, Neuroprotection and Therapeutic Strategy (BMNST Lab), Labex Medalis, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Pôle API, Ecole Supérieure de Biotechnologie, 300 Boulevard Sébastien Brant, 67412, Illkirch, France
| | - Lucas D Pham-Van
- INSERM U1119, Biopathology of Myelin, Neuroprotection and Therapeutic Strategy (BMNST Lab), Labex Medalis, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Pôle API, Ecole Supérieure de Biotechnologie, 300 Boulevard Sébastien Brant, 67412, Illkirch, France
| | - Dominique Bagnard
- INSERM U1119, Biopathology of Myelin, Neuroprotection and Therapeutic Strategy (BMNST Lab), Labex Medalis, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Pôle API, Ecole Supérieure de Biotechnologie, 300 Boulevard Sébastien Brant, 67412, Illkirch, France.
| |
Collapse
|
9
|
Zhang S, Zhang Y, Dong Y, Guo L, Zhang Z, Shao B, Qi J, Zhou H, Zhu W, Yan X, Hong G, Zhang L, Zhang X, Tang M, Zhao C, Gao X, Chai R. Knockdown of Foxg1 in supporting cells increases the trans-differentiation of supporting cells into hair cells in the neonatal mouse cochlea. Cell Mol Life Sci 2020; 77:1401-1419. [PMID: 31485717 PMCID: PMC7113235 DOI: 10.1007/s00018-019-03291-2] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 08/08/2019] [Accepted: 08/28/2019] [Indexed: 12/17/2022]
Abstract
Foxg1 is one of the forkhead box genes that are involved in morphogenesis, cell fate determination, and proliferation, and Foxg1 was previously reported to be required for morphogenesis of the mammalian inner ear. However, Foxg1 knock-out mice die at birth, and thus the role of Foxg1 in regulating hair cell (HC) regeneration after birth remains unclear. Here we used Sox2CreER/+ Foxg1loxp/loxp mice and Lgr5-EGFPCreER/+ Foxg1loxp/loxp mice to conditionally knock down Foxg1 specifically in Sox2+ SCs and Lgr5+ progenitors, respectively, in neonatal mice. We found that Foxg1 conditional knockdown (cKD) in Sox2+ SCs and Lgr5+ progenitors at postnatal day (P)1 both led to large numbers of extra HCs, especially extra inner HCs (IHCs) at P7, and these extra IHCs with normal hair bundles and synapses could survive at least to P30. The EdU assay failed to detect any EdU+ SCs, while the SC number was significantly decreased in Foxg1 cKD mice, and lineage tracing data showed that much more tdTomato+ HCs originated from Sox2+ SCs in Foxg1 cKD mice compared to the control mice. Moreover, the sphere-forming assay showed that Foxg1 cKD in Lgr5+ progenitors did not significantly change their sphere-forming ability. All these results suggest that Foxg1 cKD promotes HC regeneration and leads to large numbers of extra HCs probably by inducing direct trans-differentiation of SCs and progenitors to HCs. Real-time qPCR showed that cell cycle and Notch signaling pathways were significantly down-regulated in Foxg1 cKD mice cochlear SCs. Together, this study provides new evidence for the role of Foxg1 in regulating HC regeneration from SCs and progenitors in the neonatal mouse cochlea.
Collapse
Affiliation(s)
- Shasha Zhang
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, 210096, China
| | - Yuan Zhang
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, 210096, China
| | - Ying Dong
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, 210096, China
| | - Lingna Guo
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, 210096, China
| | - Zhong Zhang
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, 210096, China
| | - Buwei Shao
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, 210096, China
| | - Jieyu Qi
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, 210096, China
| | - Han Zhou
- Jiangsu Provincial Key Medical Discipline (Laboratory), Department of Otolaryngology Head and Neck Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Weijie Zhu
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, 210096, China
| | - Xiaoqian Yan
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, 210096, China
| | - Guodong Hong
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, 210096, China
| | - Liyan Zhang
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, 210096, China
| | - Xiaoli Zhang
- Jiangsu Provincial Key Medical Discipline (Laboratory), Department of Otolaryngology Head and Neck Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Mingliang Tang
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, 210096, China
| | - Chunjie Zhao
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, 210096, China
| | - Xia Gao
- Jiangsu Provincial Key Medical Discipline (Laboratory), Department of Otolaryngology Head and Neck Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Renjie Chai
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, 210096, China.
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Science, Beijing, China.
- Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 211189, China.
- Jiangsu Provincial Key Medical Discipline (Laboratory), Department of Otolaryngology Head and Neck Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China.
- Key Laboratory of Hearing Medicine of NHFPC, ENT Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, Shanghai Engineering Research Centre of Cochlear Implant, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200031, China.
| |
Collapse
|
10
|
Wegleiter T, Buthey K, Gonzalez-Bohorquez D, Hruzova M, Bin Imtiaz MK, Abegg A, Mebert I, Molteni A, Kollegger D, Pelczar P, Jessberger S. Palmitoylation of BMPR1a regulates neural stem cell fate. Proc Natl Acad Sci U S A 2019; 116:25688-25696. [PMID: 31772009 PMCID: PMC6926058 DOI: 10.1073/pnas.1912671116] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Neural stem cells (NSCs) generate neurons and glial cells throughout embryonic and postnatal brain development. The role of S-palmitoylation (also referred to as S-acylation), a reversible posttranslational lipid modification of proteins, in regulating the fate and activity of NSCs remains largely unknown. We used an unbiased screening approach to identify proteins that are S-acylated in mouse NSCs and showed that bone morphogenic protein receptor 1a (BMPR1a), a core mediator of BMP signaling, is palmitoylated. Genetic manipulation of S-acylated sites affects the localization and trafficking of BMPR1a and leads to altered BMP signaling. Strikingly, defective palmitoylation of BMPR1a modulates NSC function within the mouse brain, resulting in enhanced oligodendrogenesis. Thus, we identified a mechanism regulating the behavior of NSCs and provided the framework to characterize dynamic posttranslational lipid modifications of proteins in the context of NSC biology.
Collapse
Affiliation(s)
- Thomas Wegleiter
- Laboratory of Neural Plasticity, Faculties of Medicine and Science, Brain Research Institute, University of Zurich, 8057 Zurich, Switzerland;
| | - Kilian Buthey
- Laboratory of Neural Plasticity, Faculties of Medicine and Science, Brain Research Institute, University of Zurich, 8057 Zurich, Switzerland
| | - Daniel Gonzalez-Bohorquez
- Laboratory of Neural Plasticity, Faculties of Medicine and Science, Brain Research Institute, University of Zurich, 8057 Zurich, Switzerland
| | - Martina Hruzova
- Laboratory of Neural Plasticity, Faculties of Medicine and Science, Brain Research Institute, University of Zurich, 8057 Zurich, Switzerland
| | - Muhammad Khadeesh Bin Imtiaz
- Laboratory of Neural Plasticity, Faculties of Medicine and Science, Brain Research Institute, University of Zurich, 8057 Zurich, Switzerland
| | - Andrin Abegg
- Laboratory of Neural Plasticity, Faculties of Medicine and Science, Brain Research Institute, University of Zurich, 8057 Zurich, Switzerland
| | - Iliana Mebert
- Laboratory of Neural Plasticity, Faculties of Medicine and Science, Brain Research Institute, University of Zurich, 8057 Zurich, Switzerland
| | - Adriano Molteni
- Laboratory of Neural Plasticity, Faculties of Medicine and Science, Brain Research Institute, University of Zurich, 8057 Zurich, Switzerland
| | - Dominik Kollegger
- Laboratory of Neural Plasticity, Faculties of Medicine and Science, Brain Research Institute, University of Zurich, 8057 Zurich, Switzerland
| | - Pawel Pelczar
- Center for Transgenic Models, University of Basel, 4001 Basel, Switzerland
| | - Sebastian Jessberger
- Laboratory of Neural Plasticity, Faculties of Medicine and Science, Brain Research Institute, University of Zurich, 8057 Zurich, Switzerland;
| |
Collapse
|
11
|
Kang M, Lee YS. The impact of RASopathy-associated mutations on CNS development in mice and humans. Mol Brain 2019; 12:96. [PMID: 31752929 PMCID: PMC6873535 DOI: 10.1186/s13041-019-0517-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 10/28/2019] [Indexed: 01/04/2023] Open
Abstract
The RAS signaling pathway is involved in the regulation of developmental processes, including cell growth, proliferation, and differentiation, in the central nervous system (CNS). Germline mutations in the RAS signaling pathway genes are associated with a group of neurodevelopmental disorders, collectively called RASopathy, which includes neurofibromatosis type 1, Noonan syndrome, cardio-facio-cutaneous syndrome, and Costello syndrome. Most mutations associated with RASopathies increase the activity of the RAS-ERK signaling pathway, and therefore, most individuals with RASopathies share common phenotypes, such as a short stature, heart defects, facial abnormalities, and cognitive impairments, which are often accompanied by abnormal CNS development. Recent studies using mouse models of RASopathies demonstrated that particular mutations associated with each disorder disrupt CNS development in a mutation-specific manner. Here, we reviewed the recent literatures that investigated the developmental role of RASopathy-associated mutations using mutant mice, which provided insights into the specific contribution of RAS-ERK signaling molecules to CNS development and the subsequent impact on cognitive function in adult mice.
Collapse
Affiliation(s)
- Minkyung Kang
- Department of Physiology, Seoul National University College of Medicine, 103 Daehak-ro, Jongro-gu, Seoul, 03080, South Korea.,Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Yong-Seok Lee
- Department of Physiology, Seoul National University College of Medicine, 103 Daehak-ro, Jongro-gu, Seoul, 03080, South Korea. .,Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Korea. .,Neuroscience Research Institute, Seoul National University College of Medicine, 103 Daehak-ro, Jongro-gu, Seoul, 03080, South Korea.
| |
Collapse
|
12
|
JCPyV-Induced MAPK Signaling Activates Transcription Factors during Infection. Int J Mol Sci 2019; 20:ijms20194779. [PMID: 31561471 PMCID: PMC6801635 DOI: 10.3390/ijms20194779] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 09/18/2019] [Accepted: 09/23/2019] [Indexed: 02/07/2023] Open
Abstract
JC polyomavirus (JCPyV), a ubiquitous human pathogen, is the etiological agent of the fatal neurodegenerative disease progressive multifocal leukoencephalopathy (PML). Like most viruses, JCPyV infection requires the activation of host-cell signaling pathways in order to promote viral replication processes. Previous works have established the necessity of the extracellular signal-regulated kinase (ERK), the terminal core kinase of the mitogen-activated protein kinase (MAPK) cascade (MAPK-ERK) for facilitating transcription of the JCPyV genome. However, the underlying mechanisms by which the MAPK-ERK pathway becomes activated and induces viral transcription are poorly understood. Treatment of cells with siRNAs specific for Raf and MAP kinase kinase (MEK) targets proteins in the MAPK-ERK cascade, significantly reducing JCPyV infection. MEK, the dual-specificity kinase responsible for the phosphorylation of ERK, is phosphorylated at times congruent with early events in the virus infectious cycle. Moreover, a MAPK-specific signaling array revealed that transcription factors downstream of the MAPK cascade, including cMyc and SMAD4, are upregulated within infected cells. Confocal microscopy analysis demonstrated that cMyc and SMAD4 shuttle to the nucleus during infection, and nuclear localization is reduced when ERK is inhibited. These findings suggest that JCPyV induction of the MAPK-ERK pathway is mediated by Raf and MEK and leads to the activation of downstream transcription factors during infection. This study further defines the role of the MAPK cascade during JCPyV infection and the downstream signaling consequences, illuminating kinases as potential therapeutic targets for viral infection.
Collapse
|
13
|
Weinberg F, Griffin R, Fröhlich M, Heining C, Braun S, Spohr C, Iconomou M, Hollek V, Röring M, Horak P, Kreutzfeldt S, Warsow G, Hutter B, Uhrig S, Neumann O, Reuss D, Heiland DH, von Kalle C, Weichert W, Stenzinger A, Brors B, Glimm H, Fröhling S, Brummer T. Identification and characterization of a BRAF fusion oncoprotein with retained autoinhibitory domains. Oncogene 2019; 39:814-832. [PMID: 31558800 DOI: 10.1038/s41388-019-1021-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 09/09/2019] [Accepted: 09/11/2019] [Indexed: 12/14/2022]
Abstract
Fusion proteins involving the BRAF serine/threonine kinase occur in many cancers. The oncogenic potential of BRAF fusions has been attributed to the loss of critical N-terminal domains that mediate BRAF autoinhibition. We used whole-exome and RNA sequencing in a patient with glioblastoma multiforme to identify a rearrangement between TTYH3, encoding a membrane-resident, calcium-activated chloride channel, and BRAF intron 1, resulting in a TTYH3-BRAF fusion protein that retained all features essential for BRAF autoinhibition. Accordingly, the BRAF moiety of the fusion protein alone, which represents full-length BRAF without the amino acids encoded by exon 1 (BRAFΔE1), did not induce MEK/ERK phosphorylation or transformation. Likewise, neither the TTYH3 moiety of the fusion protein nor full-length TTYH3 provoked ERK pathway activity or transformation. In contrast, TTYH3-BRAF displayed increased MEK phosphorylation potential and transforming activity, which were caused by TTYH3-mediated tethering of near-full-length BRAF to the (endo)membrane system. Consistent with this mechanism, a synthetic approach, in which BRAFΔE1 was tethered to the membrane by fusing it to the cytoplasmic tail of CD8 also induced transformation. Furthermore, we demonstrate that TTYH3-BRAF signals largely independent of a functional RAS binding domain, but requires an intact BRAF dimer interface and activation loop phosphorylation sites. Cells expressing TTYH3-BRAF exhibited increased MEK/ERK signaling, which was blocked by clinically achievable concentrations of sorafenib, trametinib, and the paradox breaker PLX8394. These data provide the first example of a fully autoinhibited BRAF protein whose oncogenic potential is dictated by a distinct fusion partner and not by a structural change in BRAF itself.
Collapse
Affiliation(s)
- Florian Weinberg
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Centre for Biological Signalling Studies BIOSS, University of Freiburg, Freiburg, Germany
| | - Ricarda Griffin
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Martina Fröhlich
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg, Germany
| | - Christoph Heining
- Department of Translational Medical Oncology, NCT Dresden, Dresden, and DKFZ, Heidelberg, Germany.,University Hospital Carl Gustav Carus, Technical University Dresden, Dresden, Germany.,German Cancer Consortium (DKTK), Dresden, Germany
| | - Sandra Braun
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Centre for Biological Signalling Studies BIOSS, University of Freiburg, Freiburg, Germany
| | - Corinna Spohr
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Faculty of Biology, University of Freiburg, Freiburg, Germany.,Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg, Germany
| | - Mary Iconomou
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Viola Hollek
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Michael Röring
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Peter Horak
- Department of Translational Medical Oncology, NCT Heidelberg and DKFZ, Heidelberg, Germany.,DKTK, Heidelberg, Germany
| | - Simon Kreutzfeldt
- Department of Translational Medical Oncology, NCT Heidelberg and DKFZ, Heidelberg, Germany.,DKTK, Heidelberg, Germany
| | - Gregor Warsow
- Omics IT and Data Management Core Facility, DKFZ, Heidelberg, Germany.,Division of Theoretical Bioinformatics, DKFZ, Heidelberg, Germany
| | - Barbara Hutter
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg, Germany
| | - Sebastian Uhrig
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg, Germany.,Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Olaf Neumann
- DKTK, Heidelberg, Germany.,Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - David Reuss
- DKTK, Heidelberg, Germany.,Department of Neuropathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany.,Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
| | - Dieter Henrik Heiland
- Department of Neurosurgery, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Translational NeuroOncology Research Group, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Christof von Kalle
- Department of Translational Oncology, NCT Heidelberg and DKFZ, Heidelberg, Germany
| | - Wilko Weichert
- Institute of Pathology, Technical University Munich, Munich, Germany.,DKTK, Munich, Germany
| | - Albrecht Stenzinger
- DKTK, Heidelberg, Germany.,Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Benedikt Brors
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg, Germany.,DKTK, Heidelberg, Germany
| | - Hanno Glimm
- Department of Translational Medical Oncology, NCT Dresden, Dresden, and DKFZ, Heidelberg, Germany.,University Hospital Carl Gustav Carus, Technical University Dresden, Dresden, Germany.,German Cancer Consortium (DKTK), Dresden, Germany
| | - Stefan Fröhling
- Department of Translational Medical Oncology, NCT Heidelberg and DKFZ, Heidelberg, Germany. .,DKTK, Heidelberg, Germany.
| | - Tilman Brummer
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, Freiburg, Germany. .,Centre for Biological Signalling Studies BIOSS, University of Freiburg, Freiburg, Germany. .,Comprehensive Cancer Centre Freiburg, University of Freiburg, Freiburg, Germany. .,DKTK Partner Site Freiburg and DKFZ, Heidelberg, Germany.
| |
Collapse
|
14
|
Kim YE, Baek ST. Neurodevelopmental Aspects of RASopathies. Mol Cells 2019; 42:441-447. [PMID: 31250618 PMCID: PMC6602148 DOI: 10.14348/molcells.2019.0037] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 06/03/2019] [Accepted: 06/11/2019] [Indexed: 02/06/2023] Open
Abstract
RAS gene mutations are frequently found in one third of human cancers. Affecting approximately 1 in 1,000 newborns, germline and somatic gain-of-function mutations in the components of RAS/mitogen-activated protein kinase (RAS/MAPK) pathway has been shown to cause developmental disorders, known as RASopathies. Since RAS-MAPK pathway plays essential roles in proliferation, differentiation and migration involving developmental processes, individuals with RASopathies show abnormalities in various organ systems including central nervous system. The frequently seen neurological defects are developmental delay, macrocephaly, seizures, neurocognitive deficits, and structural malformations. Some of the defects stemmed from dysregulation of molecular and cellular processes affecting early neurodevelopmental processes. In this review, we will discuss the implications of RAS-MAPK pathway components in neurodevelopmental processes and pathogenesis of RASopathies.
Collapse
Affiliation(s)
- Ye Eun Kim
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology (POSTECH), Pohang 37673,
Korea
| | - Seung Tae Baek
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology (POSTECH), Pohang 37673,
Korea
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang 37673,
Korea
| |
Collapse
|
15
|
Zhu K, Sun J, Kang Z, Zou Z, Wu X, Wang Y, Wu G, Harris RA, Wang J. Repurposing of omeprazole for oligodendrocyte differentiation and remyelination. Brain Res 2019; 1710:33-42. [DOI: 10.1016/j.brainres.2018.12.037] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 12/21/2018] [Accepted: 12/22/2018] [Indexed: 12/20/2022]
|
16
|
Leucine-485 deletion variant of BRAF may exhibit the severe end of the clinical spectrum of CFC syndrome. J Hum Genet 2019; 64:499-504. [PMID: 30842599 DOI: 10.1038/s10038-019-0579-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 02/05/2019] [Accepted: 02/06/2019] [Indexed: 12/30/2022]
Abstract
The genotype-phenotype correlation in BRAF variant in cardio-facio-cutaneous (CFC) syndrome is not clearly defined. Here we report a case with a severe clinical phenotype and a novel BRAF variant, p.Leu485del. The present case showed severe intellectual disability, impaired awareness, hyperekplexia, involuntary movements, early onset refractory seizures, and delayed myelination on brain magnetic resonance imaging as well as a polycystic and dysplastic kidney, which are previously unreported anomalies in CFC or RAS/mitogen-activated protein kinase syndromes related to BRAF variant. CFC syndrome, especially caused by BRAF variant, should be included in the differential diagnosis of patients with developmental and epileptic encephalopathies and hyperekplexia. Furthermore, we need to keep in mind that missense variants or the deletion of Leucine-485 may be associated with severe symptoms.
Collapse
|
17
|
B-Raf deficiency impairs tumor initiation and progression in a murine breast cancer model. Oncogene 2019; 38:1324-1339. [DOI: 10.1038/s41388-018-0663-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 11/22/2018] [Accepted: 12/11/2018] [Indexed: 02/07/2023]
|
18
|
Hayashi C, Suzuki N. Heterogeneity of Oligodendrocytes and Their Precursor Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1190:53-62. [PMID: 31760638 DOI: 10.1007/978-981-32-9636-7_5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
While oligodendrocytes have been thought to be homogenous, a number of reports have indicated evidences of the heterogeneity of oligodendrocytes and their precursor cells, OPCs. Almost a century ago, Del Río Hortega found three and four types of oligodendrocytes with regions where they exist and their morphologies, respectively. Interfascicular oligodendrocytes are one of the three regional dependent types and are the most typical oligodendendroglial cells that myelinate axonal fibers in the white matter tracts. In the other two, perineuronal oligodendrocyes function as reserve cells for remyelination and regulate neuronal excitability, whereas perivascular oligodendrocytes may play a role in metabolic support of axons. Among the four morphological categories, type I and II oligodendrocytes form many myelin sheaths on small-diameter axons and specific signal is required for the myelination of small-diameter axons. Type III and IV oligodendrocytes myelinate a few number of axons/or one axon, whose diameters are large. A recent comprehensive gene expression analysis with single-cell RNA sequencing identifies six different populations in mature oligodendrocytes and only one population in OPCs. However, OPCs are not uniformed developmentally and regionally. Further, the capacity of OPC differentiation depends on the environments and conditions of the tissues. Taken together, oligodendrocytes and OPCs are diverse as the other cell types in the CNS. The orchestration of these cells with their specialized functions is critical for proper functioning of the CNS.
Collapse
Affiliation(s)
- Chikako Hayashi
- Department of Molecular and Cellular Biology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan.,Department of Biochemistry and Biophysics, Graduate School of Health Care Sciences, TMDU, Tokyo, Japan
| | - Nobuharu Suzuki
- Department of Molecular and Cellular Biology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan. .,Department of Biochemistry and Biophysics, Graduate School of Health Care Sciences, TMDU, Tokyo, Japan.
| |
Collapse
|
19
|
Pfeiffer V, Götz R, Camarero G, Heinsen H, Blum R, Rapp UR. Impaired neuronal maturation of hippocampal neural progenitor cells in mice lacking CRAF. PLoS One 2018; 13:e0192067. [PMID: 29590115 PMCID: PMC5873938 DOI: 10.1371/journal.pone.0192067] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 01/16/2018] [Indexed: 11/19/2022] Open
Abstract
RAF kinases are major constituents of the mitogen activated signaling pathway, regulating cell proliferation, differentiation and cell survival of many cell types, including neurons. In mammals, the family of RAF proteins consists of three members, ARAF, BRAF, and CRAF. Ablation of CRAF kinase in inbred mouse strains causes major developmental defects during fetal growth and embryonic or perinatal lethality. Heterozygous germline mutations in CRAF result in Noonan syndrome, which is characterized by neurocognitive impairment that may involve hippocampal physiology. The role of CRAF signaling during hippocampal development and generation of new postnatal hippocampal granule neurons has not been examined and may provide novel insight into the cause of hippocampal dysfunction in Noonan syndrome. In this study, by crossing CRAF-deficiency to CD-1 outbred mice, a CRAF mouse model was established which enabled us to investigate the interplay of neural progenitor proliferation and postmitotic differentiation during adult neurogenesis in the hippocampus. Albeit the general morphology of the hippocampus was unchanged, CRAF-deficient mice displayed smaller granule cell layer (GCL) volume at postnatal day 30 (P30). In CRAF-deficient mice a substantial number of abnormal, chromophilic, fast dividing cells were found in the subgranular zone (SGZ) and hilus of the dentate gyrus (DG), indicating that CRAF signaling contributes to hippocampal neural progenitor proliferation. CRAF-deficient neural progenitor cells showed an increased cell death rate and reduced neuronal maturation. These results indicate that CRAF function affects postmitotic neural cell differentiation and points to a critical role of CRAF-dependent growth factor signaling pathway in the postmitotic development of adult-born neurons.
Collapse
Affiliation(s)
- Verena Pfeiffer
- University of Würzburg, Institute of Anatomy and Cell Biology, Koellikerstraße 6, Würzburg, Germany
- University of Würzburg, Institute for Medical Radiation and Cell Research (MSZ), Versbacher Strasse 5, Würzburg, Germany
- * E-mail:
| | - Rudolf Götz
- University of Würzburg, Institute for Medical Radiation and Cell Research (MSZ), Versbacher Strasse 5, Würzburg, Germany
- Institute for Clinical Neurobiology, University Hospital Würzburg, Versbacher Strasse 5, Würzburg, Germany
| | - Guadelupe Camarero
- University of Würzburg, Institute for Medical Radiation and Cell Research (MSZ), Versbacher Strasse 5, Würzburg, Germany
| | - Helmut Heinsen
- University of Würzburg, Department of Psychiatry, Psychosomatics and Psychotherapy, Margarethe-Höppel-Platz 1, Würzburg, Germany
- Universidade de Sao Paulo Faculdade de Medicina, Pathology—LIM 44 Sao Paulo, SP, Brazil
| | - Robert Blum
- Institute for Clinical Neurobiology, University Hospital Würzburg, Versbacher Strasse 5, Würzburg, Germany
| | - Ulf Rüdiger Rapp
- University of Würzburg, Institute for Medical Radiation and Cell Research (MSZ), Versbacher Strasse 5, Würzburg, Germany
- Department of Lung Development and Remodeling, Max Planck Institute for Heart and Lung Research, Parkstr.1, Bad Nauheim, Germany
| |
Collapse
|
20
|
Eisenhardt AE, Sprenger A, Röring M, Herr R, Weinberg F, Köhler M, Braun S, Orth J, Diedrich B, Lanner U, Tscherwinski N, Schuster S, Dumaz N, Schmidt E, Baumeister R, Schlosser A, Dengjel J, Brummer T. Phospho-proteomic analyses of B-Raf protein complexes reveal new regulatory principles. Oncotarget 2018; 7:26628-52. [PMID: 27034005 PMCID: PMC5042004 DOI: 10.18632/oncotarget.8427] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 03/07/2016] [Indexed: 12/19/2022] Open
Abstract
B-Raf represents a critical physiological regulator of the Ras/RAF/MEK/ERK-pathway and a pharmacological target of growing clinical relevance, in particular in oncology. To understand how B-Raf itself is regulated, we combined mass spectrometry with genetic approaches to map its interactome in MCF-10A cells as well as in B-Raf deficient murine embryonic fibroblasts (MEFs) and B-Raf/Raf-1 double deficient DT40 lymphoma cells complemented with wildtype or mutant B-Raf expression vectors. Using a multi-protease digestion approach, we identified a novel ubiquitination site and provide a detailed B-Raf phospho-map. Importantly, we identify two evolutionary conserved phosphorylation clusters around T401 and S419 in the B-Raf hinge region. SILAC labelling and genetic/biochemical follow-up revealed that these clusters are phosphorylated in the contexts of oncogenic Ras, sorafenib induced Raf dimerization and in the background of the V600E mutation. We further show that the vemurafenib sensitive phosphorylation of the T401 cluster occurs in trans within a Raf dimer. Substitution of the Ser/Thr-residues of this cluster by alanine residues enhances the transforming potential of B-Raf, indicating that these phosphorylation sites suppress its signaling output. Moreover, several B-Raf phosphorylation sites, including T401 and S419, are somatically mutated in tumors, further illustrating the importance of phosphorylation for the regulation of this kinase.
Collapse
Affiliation(s)
- Anja E Eisenhardt
- Institute of Molecular Medicine and Cell Research (IMMZ), Faculty of Medicine, Albert-Ludwigs-University (ALU), Freiburg, Germany.,Institute of Biology III, Faculty of Biology, ALU, Freiburg, Germany.,Centre for Biological Systems Analysis (ZBSA), Freiburg, Germany
| | - Adrian Sprenger
- Centre for Biological Systems Analysis (ZBSA), Freiburg, Germany.,Institute for Experimental and Clinical Pharmacology and Toxicology, ALU, Freiburg, Germany.,INSERM U976 and Universitéi Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Michael Röring
- Institute of Molecular Medicine and Cell Research (IMMZ), Faculty of Medicine, Albert-Ludwigs-University (ALU), Freiburg, Germany.,Institute of Biology III, Faculty of Biology, ALU, Freiburg, Germany.,Centre for Biological Systems Analysis (ZBSA), Freiburg, Germany.,Spemann Graduate School of Biology and Medicine (SGBM), ALU, Freiburg, Germany
| | - Ricarda Herr
- Institute of Molecular Medicine and Cell Research (IMMZ), Faculty of Medicine, Albert-Ludwigs-University (ALU), Freiburg, Germany.,Institute of Biology III, Faculty of Biology, ALU, Freiburg, Germany.,Centre for Biological Systems Analysis (ZBSA), Freiburg, Germany
| | - Florian Weinberg
- Institute of Molecular Medicine and Cell Research (IMMZ), Faculty of Medicine, Albert-Ludwigs-University (ALU), Freiburg, Germany.,Institute of Biology III, Faculty of Biology, ALU, Freiburg, Germany.,Centre for Biological Systems Analysis (ZBSA), Freiburg, Germany
| | - Martin Köhler
- Institute of Molecular Medicine and Cell Research (IMMZ), Faculty of Medicine, Albert-Ludwigs-University (ALU), Freiburg, Germany.,Institute of Biology III, Faculty of Biology, ALU, Freiburg, Germany.,Centre for Biological Systems Analysis (ZBSA), Freiburg, Germany.,Spemann Graduate School of Biology and Medicine (SGBM), ALU, Freiburg, Germany
| | - Sandra Braun
- Institute of Molecular Medicine and Cell Research (IMMZ), Faculty of Medicine, Albert-Ludwigs-University (ALU), Freiburg, Germany.,Institute of Biology III, Faculty of Biology, ALU, Freiburg, Germany.,Centre for Biological Systems Analysis (ZBSA), Freiburg, Germany
| | - Joachim Orth
- Institute for Experimental and Clinical Pharmacology and Toxicology, ALU, Freiburg, Germany
| | - Britta Diedrich
- Centre for Biological Systems Analysis (ZBSA), Freiburg, Germany.,Department of Dermatology, University Medical Centre, ALU, Freiburg, Germany
| | - Ulrike Lanner
- Centre for Biological Systems Analysis (ZBSA), Freiburg, Germany
| | - Natalja Tscherwinski
- Institute of Biology III, Faculty of Biology, ALU, Freiburg, Germany.,Centre for Biological Systems Analysis (ZBSA), Freiburg, Germany
| | - Simon Schuster
- Institute of Biology III, Faculty of Biology, ALU, Freiburg, Germany.,Centre for Biological Systems Analysis (ZBSA), Freiburg, Germany
| | - Nicolas Dumaz
- INSERM U976 and Universitéi Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Enrico Schmidt
- Institute of Biology III, Faculty of Biology, ALU, Freiburg, Germany.,Centre for Biological Systems Analysis (ZBSA), Freiburg, Germany
| | - Ralf Baumeister
- Institute of Biology III, Faculty of Biology, ALU, Freiburg, Germany.,Centre for Biological Systems Analysis (ZBSA), Freiburg, Germany.,Freiburg Institute for Advanced Studies (FRIAS), ALU, Freiburg, Germany.,Centre for Biological Signalling Studies BIOSS, ALU, Freiburg, Germany
| | - Andreas Schlosser
- Institute of Biology III, Faculty of Biology, ALU, Freiburg, Germany.,Centre for Biological Systems Analysis (ZBSA), Freiburg, Germany.,Rudolf Virchow Center for Experimental Biomedicine, University of Würzburg, Würzburg, Germany
| | - Jörn Dengjel
- Centre for Biological Systems Analysis (ZBSA), Freiburg, Germany.,Department of Dermatology, University Medical Centre, ALU, Freiburg, Germany.,Freiburg Institute for Advanced Studies (FRIAS), ALU, Freiburg, Germany.,Centre for Biological Signalling Studies BIOSS, ALU, Freiburg, Germany.,Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Tilman Brummer
- Institute of Molecular Medicine and Cell Research (IMMZ), Faculty of Medicine, Albert-Ludwigs-University (ALU), Freiburg, Germany.,Institute of Biology III, Faculty of Biology, ALU, Freiburg, Germany.,Centre for Biological Systems Analysis (ZBSA), Freiburg, Germany.,Centre for Biological Signalling Studies BIOSS, ALU, Freiburg, Germany.,German Cancer Consortium (DKTK), Freiburg, Germany
| |
Collapse
|
21
|
Multipotency and therapeutic potential of NG2 cells. Biochem Pharmacol 2017; 141:42-55. [DOI: 10.1016/j.bcp.2017.05.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 05/12/2017] [Indexed: 12/20/2022]
|
22
|
Bernardo A, Giammarco ML, De Nuccio C, Ajmone-Cat MA, Visentin S, De Simone R, Minghetti L. Docosahexaenoic acid promotes oligodendrocyte differentiation via PPAR-γ signalling and prevents tumor necrosis factor-α-dependent maturational arrest. Biochim Biophys Acta Mol Cell Biol Lipids 2017. [PMID: 28647405 DOI: 10.1016/j.bbalip.2017.06.014] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Docosahexaenoic acid (DHA) is an essential omega-3 fatty acid known to be neuroprotective in several models of human diseases, including multiple sclerosis. The protective effects of DHA are largely attributed to its ability to interfere with the activity of transcription factors controlling immune and inflammatory responses, including the agonist-dependent transcription factor peroxisome proliferator-activated receptor-γ (PPAR-γ). In this study, we used primary oligodendrocyte progenitor (OP) cultures from neonatal rat brain to investigate whether DHA could influence OP maturation and directly promote myelination, as previously reported for selective PPAR-γ agonists. We show that, similarly to the selective PPAR-γ agonist pioglitazone (PGZ), DHA promotes OP maturation and counteracts the maturational arrest induced by TNF-α, used to mimic inflammatory conditions. The PPAR-γ antagonist GW9662 prevented both DHA-induced OP maturation and PPAR-γ nuclear translocation, supporting the hypothesis that DHA acts through the activation of PPAR-γ. In addition, both PGZ and DHA induced the phosphorylation of extracellular signal-regulated-kinase 1-2 (ERK1/2), in a PPAR-γ-dependent manner. ERK1/2 activity is known to regulate the transition from OPs to immature oligodendrocytes and the presence of specific inhibitors of ERK1/2 phosphorylation (U0126 or PD98059) prevented the differentiating effects of both DHA and PGZ. These results indicate that DHA might influence the process of OP maturation through its PPAR-γ agonistic activity and provide novel molecular mechanisms for the action of this dietary fatty acid, further supporting the nutritional intervention in demyelinating diseases such as multiple sclerosis.
Collapse
Affiliation(s)
- A Bernardo
- Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - M L Giammarco
- Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - C De Nuccio
- Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - M A Ajmone-Cat
- Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - S Visentin
- Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - R De Simone
- Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - L Minghetti
- Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; Research Coordination and Support Service, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy.
| |
Collapse
|
23
|
Dorard C, Vucak G, Baccarini M. Deciphering the RAS/ERK pathway in vivo. Biochem Soc Trans 2017; 45:27-36. [PMID: 28202657 DOI: 10.1042/bst20160135] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 11/03/2016] [Accepted: 11/07/2016] [Indexed: 12/19/2022]
Abstract
The RAS/ERK pathway has been intensely studied for about three decades, not least because of its role in human pathologies. ERK activation is observed in the majority of human cancers; in about one-third of them, it is driven by mutational activation of pathway components. The pathway is arguably one of the best targets for molecule-based pharmacological intervention, and several small-molecule inhibitors are in clinical use. Genetically engineered mouse models have greatly contributed to our understanding of signaling pathways in development, tissue homeostasis, and disease. In the specific case of the RAS/ERK pathway, they have revealed unique biological roles of structurally and functionally similar proteins, new kinase-independent effectors, and unsuspected relationships with other cascades. This short review summarizes the contribution of mouse models to our current understanding of the pathway.
Collapse
Affiliation(s)
- Coralie Dorard
- Max F. Perutz Laboratories, Center for Molecular Biology, University of Vienna, Vienna 1030, Austria
| | - Georg Vucak
- Max F. Perutz Laboratories, Center for Molecular Biology, University of Vienna, Vienna 1030, Austria
| | - Manuela Baccarini
- Max F. Perutz Laboratories, Center for Molecular Biology, University of Vienna, Vienna 1030, Austria
| |
Collapse
|
24
|
Akkermann R, Aprico A, Perera AA, Bujalka H, Cole AE, Xiao J, Field J, Kilpatrick TJ, Binder MD. The TAM receptor Tyro3 regulates myelination in the central nervous system. Glia 2017; 65:581-591. [PMID: 28145605 DOI: 10.1002/glia.23113] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 12/05/2016] [Accepted: 12/20/2016] [Indexed: 11/12/2022]
Abstract
Myelin is an essential component of the mammalian nervous system, facilitating rapid conduction of electrical impulses by axons, as well as providing trophic support to neurons. Within the central nervous system, the oligodendrocyte is the specialized neural cell responsible for producing myelin by a process that is thought to be regulated by both activity dependent and independent mechanisms but in incompletely understood ways. We have previously identified that the protein Gas6, a ligand for a family of tyrosine kinase receptors known as the TAM (Tyro3, Axl, and Mertk) receptors, directly increases oligodendrocyte induced myelination in vitro. Gas6 can bind to and activate all three TAM receptors, but the high level of expression of Tyro3 on oligodendrocytes makes this receptor the principal candidate for transducing the pro-myelinating effect of Gas6. In this study, we establish that in the absence of Tyro3, the pro-myelinating effect of Gas6 is lost, that developmental myelination is delayed and that the myelin produced is thinner than normal. We show that this effect is specific to the myelination process and not due to changes in the proliferation or differentiation of oligodendrocyte precursor cells. We have further demonstrated that the reduction in myelination is due to the loss of Tyro3 on oligodendrocytes, and this effect may be mediated by activation of Erk1. Collectively, our findings indicate the critical importance of Tyro3 in potentiating central nervous system myelination. GLIA 2017 GLIA 2017;65:581-591.
Collapse
Affiliation(s)
- Rainer Akkermann
- Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Andrea Aprico
- The Florey Institute of Neuroscience and Mental Health, 30 Royal Parade (Cnr Genetics Lane), Parkville, Victoria, 3052, Australia
| | - Ashwyn A Perera
- The Florey Institute of Neuroscience and Mental Health, 30 Royal Parade (Cnr Genetics Lane), Parkville, Victoria, 3052, Australia
| | - Helena Bujalka
- Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Alistair E Cole
- Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Junhua Xiao
- Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Judith Field
- Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Victoria, 3010, Australia.,The Florey Institute of Neuroscience and Mental Health, 30 Royal Parade (Cnr Genetics Lane), Parkville, Victoria, 3052, Australia
| | - Trevor J Kilpatrick
- Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Victoria, 3010, Australia.,The Florey Institute of Neuroscience and Mental Health, 30 Royal Parade (Cnr Genetics Lane), Parkville, Victoria, 3052, Australia
| | - Michele D Binder
- Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Victoria, 3010, Australia.,The Florey Institute of Neuroscience and Mental Health, 30 Royal Parade (Cnr Genetics Lane), Parkville, Victoria, 3052, Australia
| |
Collapse
|
25
|
Zhong J. RAS and downstream RAF-MEK and PI3K-AKT signaling in neuronal development, function and dysfunction. Biol Chem 2016; 397:215-22. [PMID: 26760308 DOI: 10.1515/hsz-2015-0270] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 01/04/2016] [Indexed: 12/12/2022]
Abstract
In postmitotic neurons, the activation of RAS family small GTPases regulates survival, growth and differentiation. Dysregulation of RAS or its major effector pathway, the cascade of RAF-, mitogen-activated and extracellular-signal regulated kinase kinases (MEK), and extracellular-signal regulated kinases (ERK) causes the RASopathies, a group of neurodevelopmental disorders whose pathogenic mechanisms are the subject of intense research. I here summarize the functions of RAS-RAF-MEK-ERK signaling in neurons in vivo, and discuss perspectives for harnessing this pathway to enable novel treatments for nervous system injury, the RASopathies, and possibly other neurological conditions.
Collapse
|
26
|
G protein-coupled receptor 37 is a negative regulator of oligodendrocyte differentiation and myelination. Nat Commun 2016; 7:10884. [PMID: 26961174 PMCID: PMC4792952 DOI: 10.1038/ncomms10884] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 01/29/2016] [Indexed: 12/22/2022] Open
Abstract
While the formation of myelin by oligodendrocytes is critical for the function of the
central nervous system, the molecular mechanism controlling oligodendrocyte
differentiation remains largely unknown. Here we identify G protein-coupled receptor
37 (GPR37) as an inhibitor of late-stage oligodendrocyte differentiation and
myelination. GPR37 is enriched in oligodendrocytes and its expression increases
during their differentiation into myelin forming cells. Genetic deletion of
Gpr37 does not affect the number of oligodendrocyte precursor cells, but
results in precocious oligodendrocyte differentiation and hypermyelination. The
inhibition of oligodendrocyte differentiation by GPR37 is mediated by suppression of
an exchange protein activated by cAMP (EPAC)-dependent activation of Raf-MAPK-ERK1/2
module and nuclear translocation of ERK1/2. Our data suggest that GPR37 regulates
central nervous system myelination by controlling the transition from
early-differentiated to mature oligodendrocytes. The molecular mechanism controlling oligodendrocyte differentiation is
not fully understood. Here the authors show that G protein coupled receptor 37 acts as a
negative regulator of CNS myelination, and this effect is mediated by suppression of ERK
signalling.
Collapse
|
27
|
Intracellular signaling pathway regulation of myelination and remyelination in the CNS. Exp Neurol 2016; 283:501-11. [PMID: 26957369 DOI: 10.1016/j.expneurol.2016.03.008] [Citation(s) in RCA: 159] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 02/26/2016] [Accepted: 03/04/2016] [Indexed: 12/23/2022]
Abstract
The restoration of myelin sheaths on demyelinated axons remains a major obstacle in the treatment of multiple sclerosis (MS). Currently approved therapies work by modulating the immune system to reduce the number and rate of lesion formation but are only partially effective since they are not able to restore lost myelin. In the healthy CNS, myelin continues to be generated throughout life and spontaneous remyelination occurs readily in response to insults. In patients with MS, however, remyelination eventually fails, at least in part as a result of a failure of oligodendrocyte precursor cell (OPC) differentiation and the subsequent production of new myelin. A better understanding of the molecular mechanisms and signaling pathways that drive the process of myelin sheath formation is therefore important in order to speed the development of novel therapeutics designed to target remyelination. Here we review data supporting critical roles for three highly conserved intracellular signaling pathways: Wnt/β-catenin, PI3K/AKT/mTOR, and ERK/MAPK in the regulation of OPC differentiation and myelination both during development and in remyelination. Potential points of crosstalk between the three pathways and important areas for future research are also discussed.
Collapse
|
28
|
Réus GZ, Abaleira HM, Titus SE, Arent CO, Michels M, da Luz JR, dos Santos MAB, Carlessi AS, Matias BI, Bruchchen L, Steckert AV, Ceretta LB, Dal-Pizzol F, Quevedo J. Effects of ketamine administration on the phosphorylation levels of CREB and TrKB and on oxidative damage after infusion of MEK inhibitor. Pharmacol Rep 2016; 68:177-84. [DOI: 10.1016/j.pharep.2015.08.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Revised: 07/31/2015] [Accepted: 08/13/2015] [Indexed: 12/20/2022]
|
29
|
Kremer D, Cui QL, Göttle P, Kuhlmann T, Hartung HP, Antel J, Küry P. CXCR7 Is Involved in Human Oligodendroglial Precursor Cell Maturation. PLoS One 2016; 11:e0146503. [PMID: 26741980 PMCID: PMC4704822 DOI: 10.1371/journal.pone.0146503] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 12/17/2015] [Indexed: 11/18/2022] Open
Abstract
Differentiation of oligodendroglial precursor cells (OPCs), a crucial prerequisite for central nervous system (CNS) remyelination in diseases such as Multiple Sclerosis (MS), is modulated by a multitude of extrinsic and intrinsic factors. In a previous study we revealed that the chemokine CXCL12 stimulates rodent OPC differentiation via activation of its receptor CXCR7. We could now demonstrate that CXCR7 is also expressed on NogoA- and Nkx2.2-positive oligodendroglial cells in human MS brains and that stimulation of cultured primary fetal human OPCs with CXCL12 promotes their differentiation as measured by surface marker expression and morphologic complexity. Pharmacological inhibition of CXCR7 effectively blocks these CXCL12-dependent effects. Our findings therefore suggest that a specific activation of CXCR7 could provide a means to promote oligodendroglial differentiation facilitating endogenous remyelination activities.
Collapse
Affiliation(s)
- David Kremer
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
- * E-mail:
| | - Qiao-Ling Cui
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Peter Göttle
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Tanja Kuhlmann
- Institute of Neuropathology, University Hospital Münster, Münster, Germany
| | - Hans-Peter Hartung
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Jack Antel
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Patrick Küry
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| |
Collapse
|
30
|
Affiliation(s)
- Andrea Varga
- Center of Molecular Biology, Max F. Perutz Laboratories, University of Vienna, Vienna, Austria
| | - Manuela Baccarini
- Center of Molecular Biology, Max F. Perutz Laboratories, University of Vienna, Vienna, Austria
| |
Collapse
|
31
|
Köhler M, Röring M, Schorch B, Heilmann K, Stickel N, Fiala GJ, Schmitt LC, Braun S, Ehrenfeld S, Uhl FM, Kaltenbacher T, Weinberg F, Herzog S, Zeiser R, Schamel WW, Jumaa H, Brummer T. Activation loop phosphorylation regulates B-Raf in vivo and transformation by B-Raf mutants. EMBO J 2015; 35:143-61. [PMID: 26657898 DOI: 10.15252/embj.201592097] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 10/28/2015] [Indexed: 12/19/2022] Open
Abstract
Despite being mutated in cancer and RASopathies, the role of the activation segment (AS) has not been addressed for B-Raf signaling in vivo. Here, we generated a conditional knock-in mouse allowing the expression of the B-Raf(AVKA) mutant in which the AS phosphoacceptor sites T599 and S602 are replaced by alanine residues. Surprisingly, despite producing a kinase-impaired protein, the Braf(AVKA) allele does not phenocopy the lethality of Braf-knockout or paradoxically acting knock-in alleles. However, Braf(AVKA) mice display abnormalities in the hematopoietic system, a distinct facial morphology, reduced ERK pathway activity in the brain, and an abnormal gait. This phenotype suggests that maximum B-Raf activity is required for the proper development, function, and maintenance of certain cell populations. By establishing conditional murine embryonic fibroblast cultures, we further show that MEK/ERK phosphorylation and the immediate early gene response toward growth factors are impaired in the presence of B-Raf(AVKA). Importantly, alanine substitution of T599/S602 impairs the transformation potential of oncogenic non-V600E B-Raf mutants and a fusion protein, suggesting that blocking their phosphorylation could represent an alternative strategy to ATP-competitive inhibitors.
Collapse
Affiliation(s)
- Martin Köhler
- Faculty of Medicine, Institute of Molecular Medicine and Cell Research, Albert-Ludwigs-University (ALU), Freiburg, Germany Centre for Biological Systems Analysis ZBSA, ALU, Freiburg, Germany Spemann Graduate School for Biology and Medicine, ALU, Freiburg, Germany Faculty of Biology, ALU, Freiburg, Germany
| | - Michael Röring
- Faculty of Medicine, Institute of Molecular Medicine and Cell Research, Albert-Ludwigs-University (ALU), Freiburg, Germany Centre for Biological Systems Analysis ZBSA, ALU, Freiburg, Germany Spemann Graduate School for Biology and Medicine, ALU, Freiburg, Germany Faculty of Biology, ALU, Freiburg, Germany
| | - Björn Schorch
- Centre for Biological Systems Analysis ZBSA, ALU, Freiburg, Germany Spemann Graduate School for Biology and Medicine, ALU, Freiburg, Germany Faculty of Biology, ALU, Freiburg, Germany
| | - Katharina Heilmann
- Centre for Biological Systems Analysis ZBSA, ALU, Freiburg, Germany Faculty of Biology, ALU, Freiburg, Germany
| | - Natalie Stickel
- Spemann Graduate School for Biology and Medicine, ALU, Freiburg, Germany Faculty of Biology, ALU, Freiburg, Germany Department of Hematology and Oncology, University Medical Center ALU, Freiburg, Germany
| | - Gina J Fiala
- Spemann Graduate School for Biology and Medicine, ALU, Freiburg, Germany Faculty of Biology, ALU, Freiburg, Germany Centre for Biological Signalling Studies BIOSS, ALU, Freiburg, Germany Department of Molecular Immunology, Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Lisa C Schmitt
- Faculty of Medicine, Institute of Molecular Medicine and Cell Research, Albert-Ludwigs-University (ALU), Freiburg, Germany Centre for Biological Systems Analysis ZBSA, ALU, Freiburg, Germany Faculty of Biology, ALU, Freiburg, Germany
| | - Sandra Braun
- Faculty of Medicine, Institute of Molecular Medicine and Cell Research, Albert-Ludwigs-University (ALU), Freiburg, Germany Centre for Biological Systems Analysis ZBSA, ALU, Freiburg, Germany Centre for Biological Signalling Studies BIOSS, ALU, Freiburg, Germany
| | - Sophia Ehrenfeld
- Faculty of Medicine, Institute of Molecular Medicine and Cell Research, Albert-Ludwigs-University (ALU), Freiburg, Germany Faculty of Biology, ALU, Freiburg, Germany
| | - Franziska M Uhl
- Faculty of Medicine, Institute of Molecular Medicine and Cell Research, Albert-Ludwigs-University (ALU), Freiburg, Germany Faculty of Biology, ALU, Freiburg, Germany
| | - Thorsten Kaltenbacher
- Faculty of Medicine, Institute of Molecular Medicine and Cell Research, Albert-Ludwigs-University (ALU), Freiburg, Germany Faculty of Biology, ALU, Freiburg, Germany
| | - Florian Weinberg
- Faculty of Medicine, Institute of Molecular Medicine and Cell Research, Albert-Ludwigs-University (ALU), Freiburg, Germany Centre for Biological Systems Analysis ZBSA, ALU, Freiburg, Germany Faculty of Biology, ALU, Freiburg, Germany
| | - Sebastian Herzog
- Centre for Biological Systems Analysis ZBSA, ALU, Freiburg, Germany Centre for Biological Signalling Studies BIOSS, ALU, Freiburg, Germany Department of Molecular Immunology, Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Robert Zeiser
- Department of Hematology and Oncology, University Medical Center ALU, Freiburg, Germany Centre for Biological Signalling Studies BIOSS, ALU, Freiburg, Germany Comprehensive Cancer Centre, Freiburg, Germany German Consortium for Translational Cancer Research DKTK, Standort Freiburg, Germany
| | - Wolfgang W Schamel
- Centre for Biological Signalling Studies BIOSS, ALU, Freiburg, Germany Department of Molecular Immunology, Faculty of Biology, University of Freiburg, Freiburg, Germany Center for Chronic Immunodeficiency CCI, University Medical Center, Freiburg, Germany
| | - Hassan Jumaa
- Centre for Biological Signalling Studies BIOSS, ALU, Freiburg, Germany Department of Molecular Immunology, Faculty of Biology, University of Freiburg, Freiburg, Germany Institute of Immunology, University Hospital Ulm, Ulm, Germany
| | - Tilman Brummer
- Faculty of Medicine, Institute of Molecular Medicine and Cell Research, Albert-Ludwigs-University (ALU), Freiburg, Germany Centre for Biological Systems Analysis ZBSA, ALU, Freiburg, Germany Faculty of Biology, ALU, Freiburg, Germany Centre for Biological Signalling Studies BIOSS, ALU, Freiburg, Germany Comprehensive Cancer Centre, Freiburg, Germany German Consortium for Translational Cancer Research DKTK, Standort Freiburg, Germany
| |
Collapse
|
32
|
An S, Yang Y, Ward R, Liu Y, Guo XX, Xu TR. A-Raf: A new star of the family of raf kinases. Crit Rev Biochem Mol Biol 2015; 50:520-31. [PMID: 26508523 DOI: 10.3109/10409238.2015.1102858] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The Ras-Raf-MEK-MAPK (mitogen-activated protein kinase)-signaling pathway plays a key role in the regulation of many cellular functions, including cell proliferation, differentiation and transformation, by transmitting signals from membrane receptors to various cytoplasmic and nuclear targets. One of the key components of this pathway is the serine/threonine protein kinase, Raf. The Raf family kinases (A-Raf, B-Raf and C-Raf) have been intensively studied since being identified in the early 1980s as retroviral oncogenes, especially with respect to the discovery of activating mutations of B-Raf in a large number of tumors which led to intensified efforts to develop drugs targeting Raf kinases. This also resulted in a rapid increase in our knowledge of the biological functions of the B-Raf and C-Raf isoforms, which may in turn be contrasted with the little that is known about A-Raf. The biological functions of A-Raf remain mysterious, although it appears to share some of the basic properties of the other two isoforms. Recently, emerging evidence has begun to reveal the functions of A-Raf, of which some are kinase-independent. These include the inhibition of apoptosis by binding to MST2, acting as safeguard against oncogenic transformation by suppressing extracellular signal-regulated kinases (ERK) activation and playing a role in resistance to Raf inhibitors. In this review, we discuss the regulation of A-Raf protein expression, and the roles of A-Raf in apoptosis and cancer, with a special focus on its role in resistance to Raf inhibitors. We also describe the scaffold functions of A-Raf and summarize the unexpected complexity of Raf signaling.
Collapse
Affiliation(s)
- Su An
- a Faculty of Life Science and Technology , Kunming University of Science and Technology , Kunming , Yunnan , China and
| | - Yang Yang
- a Faculty of Life Science and Technology , Kunming University of Science and Technology , Kunming , Yunnan , China and
| | - Richard Ward
- b Molecular Pharmacology Group, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow , Scotland , UK
| | - Ying Liu
- a Faculty of Life Science and Technology , Kunming University of Science and Technology , Kunming , Yunnan , China and
| | - Xiao-Xi Guo
- a Faculty of Life Science and Technology , Kunming University of Science and Technology , Kunming , Yunnan , China and
| | - Tian-Rui Xu
- a Faculty of Life Science and Technology , Kunming University of Science and Technology , Kunming , Yunnan , China and
| |
Collapse
|
33
|
Elhannan S, Taha S, Ben Khalaf N, Bakheit H, Fathallah MD, Bakhiet M. Induction of dissociated cytokine profiles by ISRAA with selective critical involvement of ERK1/2 in its signaling functions. Int J Mol Med 2015; 36:1583-92. [PMID: 26499109 DOI: 10.3892/ijmm.2015.2376] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Accepted: 09/09/2015] [Indexed: 11/05/2022] Open
Abstract
The immune system-released activating agent (ISRAA) is an immune mediator activated as a result of a nerve stimulus initiated by immune challenge. We have previously demonstrated that ISRAA and tumor necrosis factor (TNF) receptor 1 (TNFR1) share an interspecies-conserved motif (72% homology) that induces the apoptosis and proliferation of human peripheral blood mononuclear cells (hPBMCs) in a dose-dependent manner. In the present study, cytokine profiles were examined in response to the stimulation of hPBMCs with ISRAA. Furthermore, the signaling pathways induced by ISRAA were mapped. The results revealed high measurable levels of TNF-α, interleukin (IL)-6, IL-8, IL-10 and interferon (IFN)-γ, but not IL-4, IL-17 (IL-17A) or transforming growth factor (TGF)-β. The analysis of signaling pathways revealed the activation of extracellular-regulated protein kinase (ERK)1/2 as a downstream signal in the mitogen‑activated protein kinase (MAPK) pathway during TNF‑α and IL-6 production and apoptosis, but not during proliferation following stimulation with ISRAA by triggering the Fas-associated protein with death domain (FADD). STAT3 was found to be unphosphorylated in the ISRAA‑stimulated hPBMCs, and STAT3 was ubiquitously expressed in unstimulated cells, suggesting that ISRAA has a protein inhibitor of activated STAT (PIAS)-like activity, by functioning as a negative regulator of the effects of STAT3 on the Janus kinase (JAK)/STAT pathway. The determination of the nature of cytokine responses together with the signaling pathways of cellular activity induced by ISRAA paves the way for the investigation of a potential target of ISRAA and for the development of novel therapeutic approaches for the treatment of immune-regulated disorders.
Collapse
Affiliation(s)
- Sahar Elhannan
- Department of Molecular Medicine, Princess Al‑Jawhara Center for Genetics and Inherited Diseases, College of Medicine and Medical Sciences, Arabian Gulf University, Manama, Bahrain
| | - Safa Taha
- Department of Molecular Medicine, Princess Al‑Jawhara Center for Genetics and Inherited Diseases, College of Medicine and Medical Sciences, Arabian Gulf University, Manama, Bahrain
| | - Noureddine Ben Khalaf
- Department of Molecular Medicine, Princess Al‑Jawhara Center for Genetics and Inherited Diseases, College of Medicine and Medical Sciences, Arabian Gulf University, Manama, Bahrain
| | - Halla Bakheit
- Department of Molecular Medicine, Princess Al‑Jawhara Center for Genetics and Inherited Diseases, College of Medicine and Medical Sciences, Arabian Gulf University, Manama, Bahrain
| | - M Dahmani Fathallah
- Biotechnology Program, College of Graduate Studies, Arabian Gulf University, Manama, Bahrain
| | - Moiz Bakhiet
- Department of Molecular Medicine, Princess Al‑Jawhara Center for Genetics and Inherited Diseases, College of Medicine and Medical Sciences, Arabian Gulf University, Manama, Bahrain
| |
Collapse
|
34
|
Peckham H, Giuffrida L, Wood R, Gonsalvez D, Ferner A, Kilpatrick TJ, Murray SS, Xiao J. Fyn is an intermediate kinase that BDNF utilizes to promote oligodendrocyte myelination. Glia 2015; 64:255-69. [PMID: 26449489 DOI: 10.1002/glia.22927] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2015] [Revised: 08/17/2015] [Accepted: 09/15/2015] [Indexed: 01/17/2023]
Abstract
Fyn, a member of the Src family of nonreceptor tyrosine kinases, promotes central nervous system myelination during development; however the mechanisms mediating this effect remain unknown. Here we show that Fyn phosphorylation is modulated by BDNF in vivo. Concordant with this, we find that BDNF stimulates Fyn phosphorylation in myelinating cocultures, an effect dependent on oligodendroglial expression of TrkB. Importantly, PP2, a pharmacological inhibitor of Src family kinases, not only abrogated the promyelinating influence of BDNF in vitro, but also attenuated BDNF-induced phosphorylation of Erk1/2 in oligodendrocytes. Over-expression of Fyn in oligodendrocytes significantly promotes phosphorylation of Erk1/2, and promotes myelination to the extent that exogenous BDNF exerts no additive effect in vitro. In contrast, expression of a kinase-dead mutant of Fyn in oligodendrocytes significantly inhibited BDNF-induced activation of Erk1/2 and abrogated the promyelinating effect of BDNF. Analysis of white matter tracts in vivo revealed that phosphorylated Fyn primarily colocalized with mature oligodendrocytes, and was rarely observed in oligodendrocyte progenitor cells, a profile that closely parallels the detection of phosphorylated Erk1/2 in the developing central nervous system. Taken together, these data identify that Fyn kinase exerts a key role in mediating the promyelinating influence of BDNF. Here we identify a pathway in which BDNF activation of oligodendroglial TrkB receptors stimulates the phosphorylation of Fyn, a necessary step required to potentiate the phosphorylation of Erk1/2, which in turn regulates oligodendrocyte myelination.
Collapse
Affiliation(s)
- Haley Peckham
- Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, Victoria, 3010, Australia
| | - Lauren Giuffrida
- Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, Victoria, 3010, Australia
| | - Rhiannon Wood
- Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, Victoria, 3010, Australia
| | - David Gonsalvez
- Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, Victoria, 3010, Australia
| | - Anita Ferner
- Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, Victoria, 3010, Australia
| | - Trevor J Kilpatrick
- Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, Victoria, 3010, Australia
| | - Simon S Murray
- Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, Victoria, 3010, Australia
| | - Junhua Xiao
- Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, Victoria, 3010, Australia
| |
Collapse
|
35
|
Chen YH, Gianino SM, Gutmann DH. Neurofibromatosis-1 regulation of neural stem cell proliferation and multilineage differentiation operates through distinct RAS effector pathways. Genes Dev 2015; 29:1677-82. [PMID: 26272820 PMCID: PMC4561477 DOI: 10.1101/gad.261677.115] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 07/23/2015] [Indexed: 11/25/2022]
Abstract
In this study, Chen et al. used Nf1 genetically engineered mice coupled with genetic and pharmacologic inhibitor strategies to investigate the mechanisms of neurofibromin control of NSC proliferation and multilineage differentiation. They demonstrate that neurofibromin differentially controls NSC proliferation and multilineage differentiation through the selective use of the PI3K/AKT and RAF/MEK pathways, providing new insights into the roles for the RAS effector pathways in regulating brain NSC function. Neurofibromatosis type 1 (NF1) is a common neurodevelopmental disorder caused by impaired function of the neurofibromin RAS regulator. Using a combination of Nf1 genetically engineered mice and pharmacological/genetic inhibition approaches, we report that neurofibromin differentially controls neural stem cell (NSC) proliferation and multilineage differentiation through the selective use of the PI3K/AKT and RAF/MEK pathways. While PI3K/AKT governs neurofibromin-regulated NSC proliferation, multilineage differentiation is MEK-dependent. Moreover, whereas MEK-regulated multilineage differentiation requires Smad3-induced Jagged-1 expression and Notch activation, MEK/Smad3-regulated Hes1 induction is only responsible for astrocyte and neuronal differentiation. Collectively, these findings establish distinct roles for the RAS effector pathways in regulating brain NSC function.
Collapse
Affiliation(s)
- Yi-Hsien Chen
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Scott M Gianino
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - David H Gutmann
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| |
Collapse
|
36
|
Translational control of myelin basic protein expression by ERK2 MAP kinase regulates timely remyelination in the adult brain. J Neurosci 2015; 35:7850-65. [PMID: 25995471 DOI: 10.1523/jneurosci.4380-14.2015] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Successful myelin repair in the adult CNS requires the robust and timely production of myelin proteins to generate new myelin sheaths. The underlying regulatory mechanisms and complex molecular basis of myelin regeneration, however, remain poorly understood. Here, we investigate the role of ERK MAP kinase signaling in this process. Conditional deletion of Erk2 from cells of the oligodendrocyte lineage resulted in delayed remyelination following demyelinating injury to the adult mouse corpus callosum. The delayed repair occurred as a result of a specific deficit in the translation of the major myelin protein, MBP. In the absence of ERK2, activation of the ribosomal protein S6 kinase (p70S6K) and its downstream target, ribosomal protein S6 (S6RP), was impaired at a critical time when premyelinating oligodendrocytes were transitioning to mature cells capable of generating new myelin sheaths. Thus, we have described an important link between the ERK MAP kinase signaling cascade and the translational machinery specifically in remyelinating oligodendrocytes in vivo. These results suggest an important role for ERK2 in the translational control of MBP, a myelin protein that appears critical for ensuring the timely generation of new myelin sheaths following demyelinating injury in the adult CNS.
Collapse
|
37
|
Martinelli S, Stellacci E, Pannone L, D'Agostino D, Consoli F, Lissewski C, Silvano M, Cencelli G, Lepri F, Maitz S, Pauli S, Rauch A, Zampino G, Selicorni A, Melançon S, Digilio MC, Gelb BD, De Luca A, Dallapiccola B, Zenker M, Tartaglia M. Molecular Diversity and Associated Phenotypic Spectrum of Germline CBL Mutations. Hum Mutat 2015; 36:787-96. [PMID: 25952305 DOI: 10.1002/humu.22809] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 04/30/2015] [Indexed: 01/11/2023]
Abstract
Noonan syndrome (NS) is a relatively common developmental disorder with a pleomorphic phenotype. Mutations causing NS alter genes encoding proteins involved in the RAS-MAPK pathway. We and others identified Casitas B-lineage lymphoma proto-oncogene (CBL), which encodes an E3-ubiquitin ligase acting as a tumor suppressor in myeloid malignancies, as a disease gene underlying a condition clinically related to NS. Here, we further explored the spectrum of germline CBL mutations and their associated phenotype. CBL mutation scanning performed on 349 affected subjects with features overlapping NS and no mutation in NS genes allowed the identification of five different variants with pathological significance. Among them, two splice-site changes, one in-frame deletion, and one missense mutation affected the RING domain and/or the adjacent linker region, overlapping cancer-associated defects. A novel nonsense mutation generating a v-Cbl-like protein able to enhance signal flow through RAS was also identified. Genotype-phenotype correlation analysis performed on available records indicated that germline CBL mutations cause a variable phenotype characterized by a relatively high frequency of neurological features, predisposition to juvenile myelomonocytic leukemia, and low prevalence of cardiac defects, reduced growth, and cryptorchidism. Finally, we excluded a major contribution of two additional members of the CBL family, CBLB and CBLC, to NS and related disorders.
Collapse
Affiliation(s)
- Simone Martinelli
- Dipartimento di Ematologia, Oncologia e Medicina Molecolare, Istituto Superiore di Sanità, Rome, Italy
| | - Emilia Stellacci
- Dipartimento di Ematologia, Oncologia e Medicina Molecolare, Istituto Superiore di Sanità, Rome, Italy
| | - Luca Pannone
- Dipartimento di Ematologia, Oncologia e Medicina Molecolare, Istituto Superiore di Sanità, Rome, Italy.,Dipartimento di Medicina Sperimentale, Sapienza Università di Roma, Rome, Italy
| | - Daniela D'Agostino
- Department of Medical Genetics, McGill University Health Centre, Montreal Children's Hospital, Montreal, Quebec, Canada
| | - Federica Consoli
- Dipartimento di Medicina Sperimentale, Sapienza Università di Roma, Rome, Italy.,Laboratorio Mendel, Istituto di Ricovero e Cura a Carattere Scientifico-Casa Sollievo della Sofferenza, Rome, Italy
| | - Christina Lissewski
- Institute of Human Genetics, University Hospital of Magdeburg, Otto-von-Guericke-University, Magdeburg, Germany
| | - Marianna Silvano
- Dipartimento di Ematologia, Oncologia e Medicina Molecolare, Istituto Superiore di Sanità, Rome, Italy
| | - Giulia Cencelli
- Dipartimento di Ematologia, Oncologia e Medicina Molecolare, Istituto Superiore di Sanità, Rome, Italy
| | | | - Silvia Maitz
- Dipartimento di Pediatria, Genetica Clinica, Ospedale S. Gerardo, Università di Milano-Bicocca, Monza, Italy
| | - Silke Pauli
- Institute of Human Genetics, University of Göttingen, Göttingen, Germany
| | - Anita Rauch
- Institute of Medical Genetics, University of Zurich, Schlieren-Zurich, Switzerland
| | - Giuseppe Zampino
- Istituto di Clinica Pediatrica, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Angelo Selicorni
- Dipartimento di Pediatria, Genetica Clinica, Ospedale S. Gerardo, Università di Milano-Bicocca, Monza, Italy
| | - Serge Melançon
- Department of Medical Genetics, McGill University Health Centre, Montreal Children's Hospital, Montreal, Quebec, Canada
| | | | - Bruce D Gelb
- Mindich Child Health and Development Institute and Departments of Pediatrics and Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York
| | - Alessandro De Luca
- Laboratorio Mendel, Istituto di Ricovero e Cura a Carattere Scientifico-Casa Sollievo della Sofferenza, Rome, Italy
| | | | - Martin Zenker
- Institute of Human Genetics, University Hospital of Magdeburg, Otto-von-Guericke-University, Magdeburg, Germany
| | - Marco Tartaglia
- Dipartimento di Ematologia, Oncologia e Medicina Molecolare, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
38
|
Gonsalvez D, Ferner AH, Peckham H, Murray SS, Xiao J. The roles of extracellular related-kinases 1 and 2 signaling in CNS myelination. Neuropharmacology 2015; 110:586-593. [PMID: 25959068 DOI: 10.1016/j.neuropharm.2015.04.024] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 04/16/2015] [Accepted: 04/27/2015] [Indexed: 01/09/2023]
Abstract
Substantial progress has been made in identifying the intracellular signaling pathways that regulate central nervous system myelination. Recently, the mitogen activated protein kinase pathway, in particular the extracellular signal-related kinase 1 (Erk1) and Erk2, have been identified as critically important in mediating the effects of several growth factors that regulate oligodendroglial development and myelination. Here we will review the recent studies that identify the key role that Erk1/2 signaling plays in regulating oligodendroglial development, myelination and remyelination, discuss the potential mechanisms that Erk1/2 may utilize to influence myelination, and highlight some questions for further research. This article is part of the Special Issue entitled 'Oligodendrocytes in Health and Disease'.
Collapse
Affiliation(s)
- David Gonsalvez
- Department of Anatomy and Neuroscience, The University of Melbourne, Victoria 3010, Australia
| | - Anita H Ferner
- Department of Anatomy and Neuroscience, The University of Melbourne, Victoria 3010, Australia
| | - Haley Peckham
- Department of Anatomy and Neuroscience, The University of Melbourne, Victoria 3010, Australia
| | - Simon S Murray
- Department of Anatomy and Neuroscience, The University of Melbourne, Victoria 3010, Australia; The Florey Institute of Neuroscience and Mental Health Research, The University of Melbourne, Victoria 3010, Australia
| | - Junhua Xiao
- Department of Anatomy and Neuroscience, The University of Melbourne, Victoria 3010, Australia; The Florey Institute of Neuroscience and Mental Health Research, The University of Melbourne, Victoria 3010, Australia.
| |
Collapse
|
39
|
Van Hove I, Verslegers M, Hu TT, Carden M, Arckens L, Moons L. A proteomic approach to understand MMP-3-driven developmental processes in the postnatal cerebellum: Chaperonin CCT6A and MAP kinase as contributing factors. Dev Neurobiol 2015; 75:1033-48. [DOI: 10.1002/dneu.22272] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Revised: 01/16/2015] [Accepted: 01/16/2015] [Indexed: 02/06/2023]
Affiliation(s)
- Inge Van Hove
- Laboratory of Neural Circuit Development and Regeneration, Animal Physiology and Neurobiology Section; Department of Biology; KU Leuven Leuven Belgium
| | - Mieke Verslegers
- Laboratory of Neural Circuit Development and Regeneration, Animal Physiology and Neurobiology Section; Department of Biology; KU Leuven Leuven Belgium
| | - Tjing-Tjing Hu
- Laboratory of Neuroplasticity and Neuroproteomics, Animal Physiology and Neurobiology Section; Department of Biology; KU Leuven Leuven Belgium
| | - Martin Carden
- School of Biosciences, University of Kent; Canterbury CT2 7NJ United Kingdom
| | - Lutgarde Arckens
- Laboratory of Neuroplasticity and Neuroproteomics, Animal Physiology and Neurobiology Section; Department of Biology; KU Leuven Leuven Belgium
| | - Lieve Moons
- Laboratory of Neural Circuit Development and Regeneration, Animal Physiology and Neurobiology Section; Department of Biology; KU Leuven Leuven Belgium
| |
Collapse
|
40
|
"RAF" neighborhood: protein-protein interaction in the Raf/Mek/Erk pathway. FEBS Lett 2014; 588:2398-406. [PMID: 24937142 PMCID: PMC4099524 DOI: 10.1016/j.febslet.2014.06.025] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Revised: 06/05/2014] [Accepted: 06/06/2014] [Indexed: 12/19/2022]
Abstract
The Raf/Mek/Erk signaling pathway, activated downstream of Ras primarily to promote proliferation, represents the best studied of the evolutionary conserved MAPK cascades. The investigation of the pathway has continued unabated since its discovery roughly 30 years ago. In the last decade, however, the identification of unexpected in vivo functions of pathway components, as well as the discovery of Raf mutations in human cancer, the ensuing quest for inhibitors, and the efforts to understand their mechanism of action, have boosted interest tremendously. From this large body of work, protein-protein interaction has emerged as a recurrent, crucial theme. This review focuses on the role of protein complexes in the regulation of the Raf/Mek/Erk pathway and in its cross-talk with other signaling cascades. Mapping these interactions and finding a way of exploiting them for therapeutic purposes is one of the challenges of future molecule-targeted therapy.
Collapse
|
41
|
The protein tyrosine phosphatase Shp2 is required for the generation of oligodendrocyte progenitor cells and myelination in the mouse telencephalon. J Neurosci 2014; 34:3767-78. [PMID: 24599474 DOI: 10.1523/jneurosci.3515-13.2014] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The protein tyrosine phosphatase Shp2 (PTPN11) is crucial for normal brain development and has been implicated in dorsal telencephalic neuronal and astroglia cell fate decisions. However, its roles in the ventral telencephalon and during oligodendrogenesis in the telencephalon remain largely unknown. Shp2 gain-of-function (GOF) mutations are observed in Noonan syndrome, a type of RASopathy associated with multiple phenotypes, including cardiovascular, craniofacial, and neurocognitive abnormalities. To gain insight into requirements for Shp2 (LOF) and the impact of abnormal Shp2 GOF mutations, we used a Shp2 conditional mutant allele (LOF) and a cre inducible Shp2-Q79R GOF transgenic mouse in combination with Olig2(cre/+) mice to target embryonic ventral telencephalic progenitors and the oligodendrocyte lineage. In the absence of Shp2 (LOF), neuronal cell types originating from progenitors in the ventral telencephalon were generated, but oligodendrocyte progenitor cell (OPC) generation was severely impaired. Late embryonic and postnatal Shp2 cKOs showed defects in the generation of OPCs throughout the telencephalon and subsequent reductions in white matter myelination. Conversely, transgenic expression of the Shp2 GOF Noonan syndrome mutation resulted in elevated OPC numbers in the embryo and postnatal brain. Interestingly, expression of this mutation negatively influenced myelination as mice displayed abnormal myelination and fewer myelinated axons in the white matter despite elevated OPC numbers. Increased proliferating OPCs and elevated MAPK activity were also observed during oligodendrogenesis after expression of Shp2 GOF mutation. These results support the notion that appropriate Shp2 activity levels control the number as well as the differentiation of oligodendrocytes during development.
Collapse
|
42
|
O'Donovan KJ, Ma K, Guo H, Wang C, Sun F, Han SB, Kim H, Wong JK, Charron J, Zou H, Son YJ, He Z, Zhong J. B-RAF kinase drives developmental axon growth and promotes axon regeneration in the injured mature CNS. ACTA ACUST UNITED AC 2014; 211:801-14. [PMID: 24733831 PMCID: PMC4010899 DOI: 10.1084/jem.20131780] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Intraneuronal activation of B-RAF kinase is sufficient to drive the growth of peripheral axon projections and enables robust regenerative axon growth in the injured optic nerve. Activation of intrinsic growth programs that promote developmental axon growth may also facilitate axon regeneration in injured adult neurons. Here, we demonstrate that conditional activation of B-RAF kinase alone in mouse embryonic neurons is sufficient to drive the growth of long-range peripheral sensory axon projections in vivo in the absence of upstream neurotrophin signaling. We further show that activated B-RAF signaling enables robust regenerative growth of sensory axons into the spinal cord after a dorsal root crush as well as substantial axon regrowth in the crush-lesioned optic nerve. Finally, the combination of B-RAF gain-of-function and PTEN loss-of-function promotes optic nerve axon extension beyond what would be predicted for a simple additive effect. We conclude that cell-intrinsic RAF signaling is a crucial pathway promoting developmental and regenerative axon growth in the peripheral and central nervous systems.
Collapse
Affiliation(s)
- Kevin J O'Donovan
- Burke Medical Research Institute, Weill Cornell Medical College of Cornell University, White Plains, NY 10605
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Annenkov A. Receptor tyrosine kinase (RTK) signalling in the control of neural stem and progenitor cell (NSPC) development. Mol Neurobiol 2013; 49:440-71. [PMID: 23982746 DOI: 10.1007/s12035-013-8532-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Accepted: 08/09/2013] [Indexed: 01/04/2023]
Abstract
Important developmental responses are elicited in neural stem and progenitor cells (NSPC) by activation of the receptor tyrosine kinases (RTK), including the fibroblast growth factor receptors, epidermal growth factor receptor, platelet-derived growth factor receptors and insulin-like growth factor receptor (IGF1R). Signalling through these RTK is necessary and sufficient for driving a number of developmental processes in the central nervous system. Within each of the four RTK families discussed here, receptors are activated by sets of ligands that do not cross-activate receptors of the other three families, and therefore, their activation can be independently regulated by ligand availability. These RTK pathways converge on a conserved core of signalling molecules, but differences between the receptors in utilisation of signalling molecules and molecular adaptors for intracellular signal propagation become increasingly apparent. Intracellular inhibitors of RTK signalling are widely involved in the regulation of developmental signalling in NSPC and often determine developmental outcomes of RTK activation. In addition, cellular responses of NSPC to the activation of a given RTK may be significantly modulated by signal strength. Cellular propensity to respond also plays a role in developmental outcomes of RTK signalling. In combination, these mechanisms regulate the balance between NSPC maintenance and differentiation during development and in adulthood. Attribution of particular developmental responses of NSPC to specific pathways of RTK signalling becomes increasingly elucidated. Co-activation of several RTK in developing NSPC is common, and analysis of co-operation between their signalling pathways may advance knowledge of RTK role in NSPC development.
Collapse
Affiliation(s)
- Alexander Annenkov
- Bone and Joint Research Unit, William Harvey Research Institute, Bart's and The London School of Medicine, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK,
| |
Collapse
|
44
|
Ahrendsen JT, Macklin W. Signaling mechanisms regulating myelination in the central nervous system. Neurosci Bull 2013; 29:199-215. [PMID: 23558589 DOI: 10.1007/s12264-013-1322-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Accepted: 02/22/2013] [Indexed: 12/19/2022] Open
Abstract
The precise and coordinated production of myelin is essential for proper development and function of the nervous system. Diseases that disrupt myelin, including multiple sclerosis, cause significant functional disability. Current treatment aims to reduce the inflammatory component of the disease, thereby preventing damage resulting from demyelination. However, therapies are not yet available to improve natural repair processes after damage has already occurred. A thorough understanding of the signaling mechanisms that regulate myelin generation will improve our ability to enhance repair. in this review, we summarize the positive and negative regulators of myelination, focusing primarily on central nervous system myelination. Axon-derived signals, extracellular signals from both diffusible factors and the extracellular matrix, and intracellular signaling pathways within myelinating oligodendrocytes are discussed. Much is known about the positive regulators that drive myelination, while less is known about the negative regulators that shift active myelination to myelin maintenance at the appropriate time. Therefore, we also provide new data on potential negative regulators of CNS myelination.
Collapse
Affiliation(s)
- Jared T Ahrendsen
- Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
| | | |
Collapse
|
45
|
Pfeiffer V, Götz R, Xiang C, Camarero G, Braun A, Zhang Y, Blum R, Heinsen H, Nieswandt B, Rapp UR. Ablation of BRaf impairs neuronal differentiation in the postnatal hippocampus and cerebellum. PLoS One 2013; 8:e58259. [PMID: 23505473 PMCID: PMC3591433 DOI: 10.1371/journal.pone.0058259] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Accepted: 02/01/2013] [Indexed: 12/30/2022] Open
Abstract
This study focuses on the role of the kinase BRaf in postnatal brain development. Mice expressing truncated, non-functional BRaf in neural stem cell-derived brain tissue demonstrate alterations in the cerebellum, with decreased sizes and fuzzy borders of the glomeruli in the granule cell layer. In addition we observed reduced numbers and misplaced ectopic Purkinje cells that showed an altered structure of their dendritic arborizations in the hippocampus, while the overall cornus ammonis architecture appeared to be unchanged. In male mice lacking BRaf in the hippocampus the size of the granule cell layer was normal at postnatal day 12 (P12) but diminished at P21, as compared to control littermates. This defect was caused by a reduced ability of dentate gyrus progenitor cells to differentiate into NeuN positive granule cell neurons. In vitro cell culture of P0/P1 hippocampal cells revealed that BRaf deficient cells were impaired in their ability to form microtubule-associated protein 2 positive neurons. Together with the alterations in behaviour, such as autoaggression and loss of balance fitness, these observations indicate that in the absence of BRaf all neuronal cellular structures develop, but neuronal circuits in the cerebellum and hippocampus are partially disturbed besides impaired neuronal generation in both structures.
Collapse
Affiliation(s)
- Verena Pfeiffer
- Institute for Medical Radiation and Cell Research (MSZ), University of Würzburg, Würzburg, Germany
| | - Rudolf Götz
- Institute for Medical Radiation and Cell Research (MSZ), University of Würzburg, Würzburg, Germany
- Institute for Clinical Neurobiology, University Hospital Würzburg, Würzburg, Germany
| | - Chaomei Xiang
- Institute for Medical Radiation and Cell Research (MSZ), University of Würzburg, Würzburg, Germany
| | - Guadelupe Camarero
- Institute for Medical Radiation and Cell Research (MSZ), University of Würzburg, Würzburg, Germany
| | - Attila Braun
- Rudolf Virchow Centre, DFG Research Centre for Experimental Biomedicine, University Hospital Würzburg, Würzburg, Germany
| | - Yina Zhang
- Institute for Medical Radiation and Cell Research (MSZ), University of Würzburg, Würzburg, Germany
| | - Robert Blum
- Institute for Clinical Neurobiology, University Hospital Würzburg, Würzburg, Germany
| | - Helmut Heinsen
- Department of Psychiatry, Morphological Brain Research Unit, University of Würzburg, Würzburg, Germany
| | - Bernhard Nieswandt
- Rudolf Virchow Centre, DFG Research Centre for Experimental Biomedicine, University Hospital Würzburg, Würzburg, Germany
| | - Ulf R. Rapp
- Institute for Medical Radiation and Cell Research (MSZ), University of Würzburg, Würzburg, Germany
- Department of Molecular Mechanisms in Lung Cancer, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- * E-mail:
| |
Collapse
|
46
|
Bonfiglio JJ, Inda C, Senin S, Maccarrone G, Refojo D, Giacomini D, Turck CW, Holsboer F, Arzt E, Silberstein S. B-Raf and CRHR1 internalization mediate biphasic ERK1/2 activation by CRH in hippocampal HT22 Cells. Mol Endocrinol 2013; 27:491-510. [PMID: 23371389 DOI: 10.1210/me.2012-1359] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
CRH is a key regulator of neuroendocrine, autonomic, and behavioral response to stress. CRH-stimulated CRH receptor 1 (CRHR1) activates ERK1/2 depending on intracellular context. In a previous work, we demonstrated that CRH activates ERK1/2 in limbic areas of the mouse brain (hippocampus and basolateral amygdala). ERK1/2 is an essential mediator of hippocampal physiological processes including emotional behavior, synaptic plasticity, learning, and memory. To elucidate the molecular mechanisms by which CRH activates ERK1/2 in hippocampal neurons, we used the mouse hippocampal cell line HT22. We document for the first time that ERK1/2 activation in response to CRH is biphasic, involving a first cAMP- and B-Raf-dependent early phase and a second phase that critically depends on CRHR1 internalization and β-arrestin2. By means of mass-spectrometry-based screening, we identified B-Raf-associated proteins that coimmunoprecipitate with endogenous B-Raf after CRHR1 activation. Using molecular and pharmacological tools, the functional impact of selected B-Raf partners in CRH-dependent ERK1/2 activation was dissected. These results indicate that 14-3-3 proteins, protein kinase A, and Rap1, are essential for early CRH-induced ERK1/2 activation, whereas dynamin and vimentin are required for the CRHR1 internalization-dependent phase. Both phases of ERK1/2 activation depend on calcium influx and are affected by calcium/calmodulin-dependent protein kinase II inactivation. Thus, this report describes the dynamics and biphasic nature of ERK1/2 activation downstream neuronal CRHR1 and identifies several new critical components of the CRHR1 signaling machinery that selectively controls the early and late phases of ERK1/2 activation, thus providing new potential therapeutic targets for stress-related disorders.
Collapse
Affiliation(s)
- Juan J Bonfiglio
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA), CONICET, Partner Institute of the Max Planck Society, Godoy Cruz 2390, C1425FQA Buenos Aires, Argentina
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Abstract
We have defined functions of MEK in regulating gliogenesis in developing cerebral cortex using loss- and gain-of-function mouse genetics. Radial progenitors deficient in both Mek1 and Mek2 fail to transition to the gliogenic mode in late embryogenesis, and astrocyte and oligodendroglial precursors fail to appear. In exploring mechanisms, we found that the key cytokine-regulated gliogenic pathway is attenuated. Further, the Ets transcription family member Etv5/Erm is strongly regulated by MEK and Erm overexpression can rescue the gliogenic potential of Mek-deleted progenitors. Remarkably, Mek1/2-deleted mice surviving postnatally exhibit cortices almost devoid of astrocytes and oligodendroglia and exhibit neurodegeneration. Conversely, expression of constitutively active MEK1 leads to a major increase in numbers of astrocytes in the adult brain. We conclude that MEK is essential for acquisition of gliogenic competence by radial progenitors and that levels of MEK activity regulate gliogenesis in the developing cortex.
Collapse
|
48
|
Sun J, Fang Y, Chen T, Guo J, Yan J, Song S, Zhang L, Liao H. WIN55, 212-2 promotes differentiation of oligodendrocyte precursor cells and improve remyelination through regulation of the phosphorylation level of the ERK 1/2 via cannabinoid receptor 1 after stroke-induced demyelination. Brain Res 2012; 1491:225-35. [PMID: 23148948 DOI: 10.1016/j.brainres.2012.11.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2012] [Revised: 11/02/2012] [Accepted: 11/07/2012] [Indexed: 02/01/2023]
Abstract
In stroke, a common cause of neurological disability in adults is that the myelin sheaths are lost through the injury or death of mature oligodendrocytes, and the failure of remyelination may be often due to insufficient proliferation and differentiation of oligodendroglial progenitors. In the current study, we used middle cerebral artery occlusion (MCAO) to induced transient focal cerebral ischemia, and found that WIN55, 212-2 augmented actively proliferating oligodendrocytes measured by CC1 immunoreactive cells within the peri-infarct areas. To establish whether these effects were associated with changes in myelin formation, we analyzed the expression of myelin basic protein (MBP) and myelin ultrastructure. We found that WIN55, 212-2 showed more extensive remyelination than vehicle at 14 days post injection (dpi). The extracellular signal-regulated kinase/mitogen-activated protein kinase (ERK/MAPK) signaling pathway may be involved in OPCs differentiation. To determine the regulatory effect of WIN55, 212-2 post-treatment on phospho-ERK 1/2 (p-ERK 1/2) after ischemia/reperfusion, Western blot analysis was performed. We found that WIN55, 212-2 regulated the phosphorylation level of the ERK 1/2 to promote OPCs survival and differentiation. Notably, cannabinoid receptor 1 is coupled to the activation of the ERK cascade. Following rimonabant combined treatment, the effect of WIN55, 212-2 on regulating the phosphorylation level of the ERK 1/2 was reversed, and the effect of accelerated myelin formation was partially inhibited. Together, we first found that WIN55, 212-2 promoted OPCs differentiation and remyelination through regulation of the level of the p-ERK 1/2 via cannabinoid receptor 1.
Collapse
Affiliation(s)
- Jing Sun
- Neurobiology Laboratory, Jiangsu Center for Drug Screening, China Pharmaceutical University, Nanjing, PR China
| | | | | | | | | | | | | | | |
Collapse
|
49
|
ERK1/ERK2 MAPK signaling is required to increase myelin thickness independent of oligodendrocyte differentiation and initiation of myelination. J Neurosci 2012; 32:8855-64. [PMID: 22745486 DOI: 10.1523/jneurosci.0137-12.2012] [Citation(s) in RCA: 163] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Wrapping of the myelin sheath around axons by oligodendrocytes is critical for the rapid conduction of electrical signals required for the normal functioning of the CNS. Myelination is a multistep process where oligodendrocytes progress through a well coordinated differentiation program regulated by multiple extracellular growth and differentiation signals. The intracellular transduction of the extracellular signals that regulate myelination is poorly understood. Here we demonstrate a critical role for two important signaling molecules, extracelluar signal-regulated protein kinases 1 and 2 (ERK1/ERK2), downstream mediators of mitogen-activated protein kinases, in the control of CNS myelin thickness. We generated and analyzed two lines of mice lacking both ERK1/ERK2 function specifically in oligodendrocyte-lineage cells. In the absence of ERK1/ERK2 signaling NG2⁺ oligodendrocyte progenitor cells proliferated and differentiated on schedule. Mutant oligodendrocytes also ensheathed axons normally and made a few wraps of compact myelin. However, the subsequent increase in myelination that correlated myelin thickness in proportion to the axon caliber failed to occur. Furthermore, although the numbers of differentiated oligodendrocytes in the adult mutants were unchanged, they showed an inability to upregulate the transcription of major myelin genes that normally occurs during active myelination. Similarly, in vitro ERK1/ERK2-deficient oligodendrocytes differentiated normally but failed to form typical myelin-like membrane sheets. None of these effects were observed in single ERK1 or ERK2 mutants. These studies suggest that the predominant role of ERK1/ERK2 signaling in vivo is in promoting rapid myelin growth to increase its thickness, subsequent to oligodendrocyte differentiation and the initiation of myelination.
Collapse
|
50
|
O’Kusky J, Ye P. Neurodevelopmental effects of insulin-like growth factor signaling. Front Neuroendocrinol 2012; 33:230-51. [PMID: 22710100 PMCID: PMC3677055 DOI: 10.1016/j.yfrne.2012.06.002] [Citation(s) in RCA: 152] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2012] [Revised: 05/09/2012] [Accepted: 06/07/2012] [Indexed: 11/28/2022]
Abstract
Insulin-like growth factor (IGF) signaling greatly impacts the development and growth of the central nervous system (CNS). IGF-I and IGF-II, two ligands of the IGF system, exert a wide variety of actions both during development and in adulthood, promoting the survival and proliferation of neural cells. The IGFs also influence the growth and maturation of neural cells, augmenting dendritic growth and spine formation, axon outgrowth, synaptogenesis, and myelination. Specific IGF actions, however, likely depend on cell type, developmental stage, and local microenvironmental milieu within the brain. Emerging research also indicates that alterations in IGF signaling likely contribute to the pathogenesis of some neurological disorders. This review summarizes experimental studies and shed light on the critical roles of IGF signaling, as well as its mechanisms, during CNS development.
Collapse
Affiliation(s)
- John O’Kusky
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada V5Z 1M9
| | - Ping Ye
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| |
Collapse
|