1
|
Chang H, Ng C, Chen Y, Wang Y, Yu I, Lee LJ, Lee L, Lee K. Elevated reactive aggression in forebrain-specific Ccn2 knockout mice. J Cell Commun Signal 2024; 18:e12040. [PMID: 39524137 PMCID: PMC11544641 DOI: 10.1002/ccs3.12040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 05/27/2024] [Accepted: 05/28/2024] [Indexed: 11/16/2024] Open
Abstract
Cellular communication network factor 2 (CCN2) is a matricellular protein that plays important roles in connective tissue. CCN2 is also expressed in the nervous system; however, its role is still unclear. To explore CCN2 function in the brain, we generated forebrain-specific Ccn2 knockout (FbCcn2 KO) mice. In this study, we examined the behavioral phenotypes of FbCcn2KO mice. Male mice lacking CCN2 in the forebrain exhibited normal locomotion, sensorimotor gating, and social behaviors but signs of anxiety and elevated reactive aggression. We checked the c-fos expression in aggression-related brain regions following the resident-intruder task (RIT), an aggression test. RIT-induced c-fos levels in the medial amygdala (MeA) were higher in FbCcn2 -/- mice as compared to controls. However, in the prefrontal cortex, RIT-induced c-fos levels in FbCcn2 -/- mice were lower than controls. Our results suggested in male mice lacking CCN2 in the olfaction-related regions, olfactory social cues elicit greater signals in the MeA, resulting in greater reactive aggression in the RIT. Further, lacking CCN2 in the prefrontal cortex, the major area related to inhibitory control and emotion regulation, may lead to signs of anxiety and the failure to suppress aggressive behaviors. Our model is useful in elaborating the mechanism underlying reactive aggression and therapeutic strategies.
Collapse
Affiliation(s)
- Ho‐Ching Chang
- College of MedicineGraduate Institute of Anatomy and Cell BiologyNational Taiwan UniversityTaipeiTaiwan
| | - Chi‐Hou Ng
- College of MedicineGraduate Institute of Anatomy and Cell BiologyNational Taiwan UniversityTaipeiTaiwan
| | - Yu‐Fu Chen
- Department of NeurologyChang Gung Memorial HospitalKeelung BranchKeelungTaiwan
| | - Yu‐Chun Wang
- Department of Otolaryngology, Head and Neck SurgeryChi‐Mei Medical CenterTainanTaiwan
| | - I‐Shing Yu
- Laboratory Animal CenterCollege of MedicineNational Taiwan UniversityTaipeiTaiwan
| | - Lukas Jyuhn‐Hsiarn Lee
- National Institute of Environmental Health SciencesNational Health Research InstitutesMiaoliTaiwan
| | - Li‐Jen Lee
- College of MedicineGraduate Institute of Anatomy and Cell BiologyNational Taiwan UniversityTaipeiTaiwan
- College of MedicineInstitute of Brain and Mind SciencesNational Taiwan UniversityTaipeiTaiwan
- Neurobiology and Cognitive Science CenterNational Taiwan UniversityTaipeiTaiwan
| | - Kuang‐Yung Lee
- Department of NeurologyChang Gung Memorial HospitalKeelung BranchKeelungTaiwan
- College of MedicineChang Gung UniversityTaoyuanTaiwan
| |
Collapse
|
2
|
Khudayberdiev S, Weiss K, Heinze A, Colombaretti D, Trausch N, Linne U, Rust MB. The actin-binding protein CAP1 represses MRTF-SRF-dependent gene expression in mouse cerebral cortex. Sci Signal 2024; 17:eadj0032. [PMID: 38713765 DOI: 10.1126/scisignal.adj0032] [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] [Received: 05/31/2023] [Accepted: 04/15/2024] [Indexed: 05/09/2024]
Abstract
Serum response factor (SRF) is an essential transcription factor for brain development and function. Here, we explored how an SRF cofactor, the actin monomer-sensing myocardin-related transcription factor MRTF, is regulated in mouse cortical neurons. We found that MRTF-dependent SRF activity in vitro and in vivo was repressed by cyclase-associated protein CAP1. Inactivation of the actin-binding protein CAP1 reduced the amount of actin monomers in the cytoplasm, which promoted nuclear MRTF translocation and MRTF-SRF activation. This function was independent of cofilin1 and actin-depolymerizing factor, and CAP1 loss of function in cortical neurons was not compensated by endogenous CAP2. Transcriptomic and proteomic analyses of cerebral cortex lysates from wild-type and Cap1 knockout mice supported the role of CAP1 in repressing MRTF-SRF-dependent signaling in vivo. Bioinformatic analysis identified likely MRTF-SRF target genes, which aligned with the transcriptomic and proteomic results. Together with our previous studies that implicated CAP1 in axonal growth cone function as well as the morphology and plasticity of excitatory synapses, our findings establish CAP1 as a crucial actin regulator in the brain relevant for formation of neuronal networks.
Collapse
Affiliation(s)
- Sharof Khudayberdiev
- Molecular Neurobiology Group, Institute of Physiological Chemistry, Philipps-University of Marburg, 35032 Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus-Liebig-University Giessen, Hans-Meerwein-Strasse 6, 35032 Marburg, Germany
| | - Kerstin Weiss
- Molecular Neurobiology Group, Institute of Physiological Chemistry, Philipps-University of Marburg, 35032 Marburg, Germany
| | - Anika Heinze
- Molecular Neurobiology Group, Institute of Physiological Chemistry, Philipps-University of Marburg, 35032 Marburg, Germany
| | - Dalila Colombaretti
- Molecular Neurobiology Group, Institute of Physiological Chemistry, Philipps-University of Marburg, 35032 Marburg, Germany
| | - Nathan Trausch
- Molecular Neurobiology Group, Institute of Physiological Chemistry, Philipps-University of Marburg, 35032 Marburg, Germany
| | - Uwe Linne
- Department of Chemistry, Philipps-University Marburg, 35032 Marburg, Germany
| | - Marco B Rust
- Molecular Neurobiology Group, Institute of Physiological Chemistry, Philipps-University of Marburg, 35032 Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus-Liebig-University Giessen, Hans-Meerwein-Strasse 6, 35032 Marburg, Germany
| |
Collapse
|
3
|
Iram T, Garcia MA, Amand J, Kaur A, Atkins M, Iyer M, Lam M, Ambiel N, Jorgens DM, Keller A, Wyss-Coray T, Kern F, Zuchero JB. SRF transcriptionally regulates the oligodendrocyte cytoskeleton during CNS myelination. Proc Natl Acad Sci U S A 2024; 121:e2307250121. [PMID: 38483990 PMCID: PMC10962977 DOI: 10.1073/pnas.2307250121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 02/10/2024] [Indexed: 03/19/2024] Open
Abstract
Myelination of neuronal axons is essential for nervous system development. Myelination requires dramatic cytoskeletal dynamics in oligodendrocytes, but how actin is regulated during myelination is poorly understood. We recently identified serum response factor (SRF)-a transcription factor known to regulate expression of actin and actin regulators in other cell types-as a critical driver of myelination in the aged brain. Yet, a major gap remains in understanding the mechanistic role of SRF in oligodendrocyte lineage cells. Here, we show that SRF is required cell autonomously in oligodendrocytes for myelination during development. Combining ChIP-seq with RNA-seq identifies SRF-target genes in oligodendrocyte precursor cells and oligodendrocytes that include actin and other key cytoskeletal genes. Accordingly, SRF knockout oligodendrocytes exhibit dramatically reduced actin filament levels early in differentiation, consistent with its role in actin-dependent myelin sheath initiation. Surprisingly, oligodendrocyte-restricted loss of SRF results in upregulation of gene signatures associated with aging and neurodegenerative diseases. Together, our findings identify SRF as a transcriptional regulator that controls the expression of cytoskeletal genes required in oligodendrocytes for myelination. This study identifies an essential pathway regulating oligodendrocyte biology with high relevance to brain development, aging, and disease.
Collapse
Affiliation(s)
- Tal Iram
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA94305
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA94305
| | - Miguel A. Garcia
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA94305
| | - Jérémy Amand
- Department of Clinical Bioinformatics, Helmholtz Institute for Pharmaceutical Research Saarland–Helmholtz Centre for Infection Research, Saarland University Campus, Saarbrücken66123, Germany
- Clinical Bioinformatics, Saarland University, Saarbrücken66123, Germany
| | - Achint Kaur
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA94305
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA94305
| | - Micaiah Atkins
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA94305
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA94305
| | - Manasi Iyer
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA94305
| | - Mable Lam
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA94305
| | - Nicholas Ambiel
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA94305
| | | | - Andreas Keller
- Department of Clinical Bioinformatics, Helmholtz Institute for Pharmaceutical Research Saarland–Helmholtz Centre for Infection Research, Saarland University Campus, Saarbrücken66123, Germany
- Clinical Bioinformatics, Saarland University, Saarbrücken66123, Germany
| | - Tony Wyss-Coray
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA94305
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA94305
| | - Fabian Kern
- Department of Clinical Bioinformatics, Helmholtz Institute for Pharmaceutical Research Saarland–Helmholtz Centre for Infection Research, Saarland University Campus, Saarbrücken66123, Germany
- Clinical Bioinformatics, Saarland University, Saarbrücken66123, Germany
| | - J. Bradley Zuchero
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA94305
| |
Collapse
|
4
|
Gao L, Zhang C, Zhu Y, Zhang N, Zhang C, Zhou S, Feng G, Huang F, Zhang L. Serum response factor promoting axonal regeneration by activating the Ras-Raf-Cofilin signaling pathway after the spinal cord injury. CNS Neurosci Ther 2024; 30:e14585. [PMID: 38421133 PMCID: PMC10851317 DOI: 10.1111/cns.14585] [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: 06/05/2023] [Revised: 11/29/2023] [Accepted: 12/19/2023] [Indexed: 03/02/2024] Open
Abstract
INTRODUCTION Serum response factor (SRF) is important in muscle development, tissue repair, and neuronal regulation. OBJECTIVES This research aims to thoroughly examine the effects of SRF on spinal cord injury (SCI) and its ability to significantly impact the recovery and regeneration of neuronal axons. METHODS The researchers created rat models of SCI and scratch injury to primary spinal cord neurons to observe the expression of relevant factors after neuronal injury. RESULTS We found that the SRF, Ras, Raf, and cofilin levels increased after injury and gradually returned to normal levels. Afterward, researchers gave rats with SCI an SRF inhibitor (CCG1423) and studied the effects with nuclear magnetic resonance and transmission electron microscopy. The SRF inhibitor rodents had worse spinal cord recovery and axon regrowth than the control group. And the apoptosis of primary neurons after scratch injury was significantly higher in the SRF inhibitor group. Additionally, the researchers utilized lentiviral transfection to modify the SRF expression in neurons. SRF overexpression increased neuron migration while silencing SRF decreased it. Finally, Western blotting and RT-PCR were conducted to examine the expression changes of related factors upon altering SRF expression. The results revealed SRF overexpression increased Ras, Raf, and cofilin expression. Silencing SRF decreased Ras, Raf, and Cofilin expression. CONCLUSION Based on our research, the SRF promotes axonal regeneration by activating the "Ras-Raf-Cofilin" signaling pathway.
Collapse
Affiliation(s)
- Limin Gao
- Institute of Neurobiology, Binzhou Medical UniversityYantaiShandong ProvinceChina
- Department of NeurobiologySchool of Basic Medical Sciences, Capital Medical UniversityBeijingChina
| | - Chen Zhang
- Institute of Neurobiology, Binzhou Medical UniversityYantaiShandong ProvinceChina
- Experimental Neurosurgery, Department of NeurosurgeryNeuroscience Center, Frankfurt University HospitalFrankfurt am MainGermany
| | - Yonglin Zhu
- Department of Bone and JointYantai Affiliated Hospital of Binzhou Medical UniversityYantaiShandongChina
| | - Naili Zhang
- Institute of Neurobiology, Binzhou Medical UniversityYantaiShandong ProvinceChina
| | - Chunlei Zhang
- Institute of Neurobiology, Binzhou Medical UniversityYantaiShandong ProvinceChina
| | - Shuai Zhou
- Institute of Neurobiology, Binzhou Medical UniversityYantaiShandong ProvinceChina
| | - Guoying Feng
- Institute of Neurobiology, Binzhou Medical UniversityYantaiShandong ProvinceChina
| | - Fei Huang
- Institute of Neurobiology, Binzhou Medical UniversityYantaiShandong ProvinceChina
- University of Health and Rehabilitation SciencesQingdaoShandong ProvinceChina
| | - Luping Zhang
- Institute of Neurobiology, Binzhou Medical UniversityYantaiShandong ProvinceChina
| |
Collapse
|
5
|
Itoh K, Ossipova O, Matsuda M, Sokol SY. Myocardin-related transcription factors regulate morphogenetic events in vertebrate embryos by controlling F-actin organization and apical constriction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.27.559818. [PMID: 37808688 PMCID: PMC10557707 DOI: 10.1101/2023.09.27.559818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Myocardin-related transcription factors (Mrtfa and Mrtfb), also known as megakaryoblastic leukemia proteins (Mkl1/MAL and Mkl2), associate with serum response factor (Srf) to regulate transcription in response to actin dynamics, however, the functions of Mrtfs in early vertebrate embryos remain largely unknown. Here we document the requirement of Mrtfs for blastopore closure at gastrulation and neural plate folding in Xenopus early embryos. Both stimulation and inhibition of Mrtf activity caused similar gross morphological phenotypes, yet the effects on F-actin distribution and cell behavior were different. Suppressing Mrtf-dependent transcription reduced overall F-actin levels and inhibited apical constriction during gastrulation and neurulation. By contrast, constitutively active Mrtf caused tricellular junction remodeling and induced apical constriction in superficial ectoderm. The underlying mechanism appeared distinct from the one utilized by known apical constriction inducers. We propose that the regulation of apical constriction is among the primary cellular responses to Mrtf. Our findings highlight a dedicated role of specific transcription factors, Mrtfs, in early morphogenetic processes.
Collapse
|
6
|
Huang H, Jing B, Zhu F, Jiang W, Tang P, Shi L, Chen H, Ren G, Xia S, Wang L, Cui Y, Yang Z, Platero AJ, Hutchins AP, Chen M, Worley PF, Xiao B. Disruption of neuronal RHEB signaling impairs oligodendrocyte differentiation and myelination through mTORC1-DLK1 axis. Cell Rep 2023; 42:112801. [PMID: 37463107 PMCID: PMC11849431 DOI: 10.1016/j.celrep.2023.112801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 05/12/2023] [Accepted: 06/26/2023] [Indexed: 07/20/2023] Open
Abstract
How neuronal signaling affects brain myelination remains poorly understood. We show dysregulated neuronal RHEB-mTORC1-DLK1 axis impairs brain myelination. Neuronal Rheb cKO impairs oligodendrocyte differentiation/myelination, with activated neuronal expression of the imprinted gene Dlk1. Neuronal Dlk1 cKO ameliorates myelination deficit in neuronal Rheb cKO mice, indicating that activated neuronal Dlk1 expression contributes to impaired myelination caused by Rheb cKO. The effect of Rheb cKO on Dlk1 expression is mediated by mTORC1; neuronal mTor cKO and Raptor cKO and pharmacological inhibition of mTORC1 recapitulate elevated neuronal Dlk1 expression. We demonstrate that both a secreted form of DLK1 and a membrane-bound DLK1 inhibit the differentiation of cultured oligodendrocyte precursor cells into oligodendrocytes expressing myelin proteins. Finally, neuronal expression of Dlk1 in transgenic mice reduces the formation of mature oligodendrocytes and myelination. This study identifies Dlk1 as an inhibitor of oligodendrocyte myelination and a mechanism linking altered neuronal signaling with oligodendrocyte dysfunction.
Collapse
Affiliation(s)
- Haijiao Huang
- Departments of Neuroscience and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen Key Laboratory for Gene Regulation and Systems Biology, Shenzhen 518055, People's Republic of China
| | - Bo Jing
- Departments of Neuroscience and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen Key Laboratory for Gene Regulation and Systems Biology, Shenzhen 518055, People's Republic of China.
| | - Feiyan Zhu
- Departments of Neuroscience and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen Key Laboratory for Gene Regulation and Systems Biology, Shenzhen 518055, People's Republic of China
| | - Wanxiang Jiang
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Ping Tang
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Liyang Shi
- Departments of Neuroscience and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen Key Laboratory for Gene Regulation and Systems Biology, Shenzhen 518055, People's Republic of China
| | - Huiting Chen
- Departments of Neuroscience and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen Key Laboratory for Gene Regulation and Systems Biology, Shenzhen 518055, People's Republic of China
| | - Guoru Ren
- Departments of Neuroscience and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen Key Laboratory for Gene Regulation and Systems Biology, Shenzhen 518055, People's Republic of China
| | - Shiyao Xia
- Departments of Neuroscience and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen Key Laboratory for Gene Regulation and Systems Biology, Shenzhen 518055, People's Republic of China
| | - Luoling Wang
- Departments of Neuroscience and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen Key Laboratory for Gene Regulation and Systems Biology, Shenzhen 518055, People's Republic of China
| | - Yiyuan Cui
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Zhiwen Yang
- Departments of Neuroscience and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen Key Laboratory for Gene Regulation and Systems Biology, Shenzhen 518055, People's Republic of China
| | - Alexander J Platero
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Andrew P Hutchins
- Departments of Neuroscience and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen Key Laboratory for Gene Regulation and Systems Biology, Shenzhen 518055, People's Republic of China
| | - Mina Chen
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Paul F Worley
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Bo Xiao
- Departments of Neuroscience and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen Key Laboratory for Gene Regulation and Systems Biology, Shenzhen 518055, People's Republic of China.
| |
Collapse
|
7
|
Reinehr S, Girbig RM, Schulte KK, Theile J, Asaad MA, Fuchshofer R, Dick H, Joachim SC. Enhanced glaucomatous damage accompanied by glial response in a new multifactorial mouse model. Front Immunol 2023; 13:1017076. [PMID: 36733392 PMCID: PMC9887307 DOI: 10.3389/fimmu.2022.1017076] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 12/19/2022] [Indexed: 01/18/2023] Open
Abstract
Introduction Glaucoma is a complex, multifactorial neurodegenerative disease, which can lead to blindness if left untreated. It seems that, among others, immune processes, elevated intraocular pressure (IOP), or a combination of these factors are responsible for glaucomatous damage. Here, we combined two glaucoma models to examine if a combination of risk factors (IOP and immune response) results in a more severe damage of retinal ganglion cells (RGCs) and the optic nerves as well as an additional glia activation. Methods Six-week-old wildtype (WT+ONA) and βB1-Connective Tissue Growth Factor (CTGF) mice (CTGF+ONA) were immunized with 1 mg ONA (optic nerve antigen). A WT and a CTGF control group (CTGF) received sodium chloride instead. IOP was measured before and every two weeks after immunization. After six weeks, electroretinogram (ERG) measurements were performed. Then, retinae and optic nerves were processed for (immuno-) histology. Further, mRNA levels of corresponding genes in optic nerve and retina were analyzed via RT-qPCR. Results Six weeks after immunization, the IOP in CTGF and CTGF+ONA mice was increased. The optic nerve of CTGF+ONA animals displayed the most severe cell inflammation, demyelination, and macroglia activation. Fewer numbers of oligodendrocytes were only observed in WT+ONA optic nerves, while more apoptotic cells triggered by the extrinsic pathway could be revealed in all three glaucoma groups. The number of microglia/macrophages was not altered within the optic nerves of all groups. The loss of neuronal cells, especially RGCs was most pronounced in CTGF+ONA retinae in the central part and this was accompanied by an enhanced activation of microglia/macrophages. Also, Müller cell activation could be noted in CTGF and CTGF+ONA retinae. Discussion In this new model, an additive degeneration could be noted in optic nerves as well as in the number of RGCs. These results suggest a potential additive role of high IOP and immune factors in glaucoma development, which will aid for understanding this multifactorial disease more precisely in the future.
Collapse
Affiliation(s)
- Sabrina Reinehr
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany,*Correspondence: Sabrina Reinehr,
| | - Renée M. Girbig
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Kim K. Schulte
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Janine Theile
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| | - M. Ali Asaad
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Rudolf Fuchshofer
- Institute of Human Anatomy and Embryology, University Regensburg, Regensburg, Germany
| | - H. Burkhard Dick
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Stephanie C. Joachim
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| |
Collapse
|
8
|
Tabuchi A, Ihara D. SRF in Neurochemistry: Overview of Recent Advances in Research on the Nervous System. Neurochem Res 2022; 47:2545-2557. [PMID: 35668335 DOI: 10.1007/s11064-022-03632-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 04/20/2022] [Accepted: 05/07/2022] [Indexed: 10/18/2022]
Abstract
Serum response factor (SRF) is a representative transcription factor that plays crucial roles in various biological phenomena by regulating immediate early genes (IEGs) and genes related to cell morphology and motility, among others. Over the years, the signal transduction pathways activating SRF have been clarified and SRF-target genes have been identified. In this overview, we initially briefly summarize the basic biology of SRF and its cofactors, ternary complex factor (TCF) and megakaryoblastic leukemia (MKL)/myocardin-related transcription factor (MRTF). Progress in the generation of nervous system-specific knockout (KO) or genetically modified mice as well as genetic analyses over the last few decades has not only identified novel SRF-target genes but also highlighted the neurochemical importance of SRF and its cofactors. Therefore, here we next present the phenotypes of mice with nervous system-specific KO of SRF or its cofactors by depicting recent findings associated with brain development, plasticity, epilepsy, stress response, and drug addiction, all of which result from function or dysfunction of the SRF axis. Last, we develop a hypothesis regarding the possible involvement of SRF and its cofactors in human neurological disorders including neurodegenerative, psychiatric, and neurodevelopmental diseases. This overview should deepen our understanding, highlight promising future directions for developing novel therapeutic strategies, and lead to illumination of the mechanisms underlying higher brain functions based on neuronal structure and function.
Collapse
Affiliation(s)
- Akiko Tabuchi
- Laboratory of Molecular Neurobiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan.
| | - Daisuke Ihara
- Laboratory of Molecular Neurobiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| |
Collapse
|
9
|
Can Demirdöğen B, Kılıç OO, Karagülle EN, Kalmaz LM, Mungan S. Single nucleotide variants around the connective tissue growth factor (CTGF/CCN2) gene and their association with multiple sclerosis risk, disability scores, and rate of disease progression. Neurol Sci 2022; 43:3867-3877. [PMID: 35091888 DOI: 10.1007/s10072-021-05852-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 12/27/2021] [Indexed: 11/24/2022]
Abstract
BACKGROUND This study aimed to explore the possible association of single nucleotide polymorphisms (SNPs) in the upstream (rs9402373) and downstream regions (rs9399005 and rs12526196) of the gene encoding connective tissue growth factor (CTGF/CCN2) with relapsing-remitting multiple sclerosis (RRMS) risk and clinical parameters including disability scores and rate of disability progression. MATERIALS AND METHODS In total, 200 patients with RRMS and 305 controls were genotyped using real-time PCR (rs1252696 C/T and rs9402373 G/C) or PCR-RFLP (rs9399005 C/T) methods. Furthermore, the association between these genotypes and clinical parameters including Expanded Disability Status Scale (EDSS) score, Multiple Sclerosis Severity Score (MSSS), age at onset, duration of disease, duration of treatment, and presence of contrast-enhancing lesions was analyzed. RESULTS rs9399005 genotypes TT and CT in the dominant model were significant predictors of RRMS vs. control status by logistic regression analysis (OR = 1.45, 95% CI = 1.01-2.08, P = .04). Moreover, these genotypes for rs9399005 were associated with a MSSS ≥ 2.4 (OR = 3.54, 95% CI = 1.56-8.05, P = .003). In addition, MSSS was lower in patients who had at least one rs12526196C allele than in the corresponding patients with the TT genotype (P = .02). CONCLUSION To our knowledge, this is the first evidence of the involvement of variants around the CTGF gene in MS risk and disability progression.
Collapse
Affiliation(s)
- Birsen Can Demirdöğen
- Department of Biomedical Engineering, TOBB University of Economics and Technology, Söğütözü, 06560, Ankara, Turkey.
| | - Osman Oğuzhan Kılıç
- Department of Biomedical Engineering, TOBB University of Economics and Technology, Söğütözü, 06560, Ankara, Turkey
| | - Elif Naz Karagülle
- Department of Biomedical Engineering, TOBB University of Economics and Technology, Söğütözü, 06560, Ankara, Turkey
| | - Latife Mekselina Kalmaz
- Department of Biomedical Engineering, TOBB University of Economics and Technology, Söğütözü, 06560, Ankara, Turkey
| | - Semra Mungan
- Neurology Clinic, Ankara City Hospital, Ankara, Turkey
| |
Collapse
|
10
|
Fu M, Peng D, Lan T, Wei Y, Wei X. Multifunctional regulatory protein connective tissue growth factor (CTGF): A potential therapeutic target for diverse diseases. Acta Pharm Sin B 2022; 12:1740-1760. [PMID: 35847511 PMCID: PMC9279711 DOI: 10.1016/j.apsb.2022.01.007] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/22/2021] [Accepted: 12/16/2021] [Indexed: 12/24/2022] Open
Abstract
Connective tissue growth factor (CTGF), a multifunctional protein of the CCN family, regulates cell proliferation, differentiation, adhesion, and a variety of other biological processes. It is involved in the disease-related pathways such as the Hippo pathway, p53 and nuclear factor kappa-B (NF-κB) pathways and thus contributes to the developments of inflammation, fibrosis, cancer and other diseases as a downstream effector. Therefore, CTGF might be a potential therapeutic target for treating various diseases. In recent years, the research on the potential of CTGF in the treatment of diseases has also been paid more attention. Several drugs targeting CTGF (monoclonal antibodies FG3149 and FG3019) are being assessed by clinical or preclinical trials and have shown promising outcomes. In this review, the cellular events regulated by CTGF, and the relationships between CTGF and pathogenesis of diseases are systematically summarized. In addition, we highlight the current researches, focusing on the preclinical and clinical trials concerned with CTGF as the therapeutic target.
Collapse
|
11
|
Yu Z, Yang Z, Ren G, Wang Y, Luo X, Zhu F, Yu S, Jia L, Chen M, Worley PF, Xiao B. GATOR2 complex-mediated amino acid signaling regulates brain myelination. Proc Natl Acad Sci U S A 2022; 119:e2110917119. [PMID: 35022234 PMCID: PMC8784133 DOI: 10.1073/pnas.2110917119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Accepted: 11/16/2021] [Indexed: 02/05/2023] Open
Abstract
Amino acids are essential for cell growth and metabolism. Amino acid and growth factor signaling pathways coordinately regulate the mechanistic target of rapamycin complex 1 (mTORC1) kinase in cell growth and organ development. While major components of amino acid signaling mechanisms have been identified, their biological functions in organ development are unclear. We aimed to understand the functions of the critically positioned amino acid signaling complex GAP activity towards Rags 2 (GATOR2) in brain development. GATOR2 mediates amino acid signaling to mTORC1 by directly linking the amino acid sensors for arginine and leucine to downstream signaling complexes. Now, we report a role of GATOR2 in oligodendrocyte myelination in postnatal brain development. We show that the disruption of GATOR2 complex by genetic deletion of meiosis regulator for oocyte development (Mios, encoding a component of GATOR2) selectively impairs the formation of myelinating oligodendrocytes, thus brain myelination, without apparent effects on the formation of neurons and astrocytes. The loss of Mios impairs cell cycle progression of oligodendrocyte precursor cells, leading to their reduced proliferation and differentiation. Mios deletion manifests a cell type-dependent effect on mTORC1 in the brain, with oligodendroglial mTORC1 selectively affected. However, the role of Mios/GATOR2 in oligodendrocyte formation and myelination involves mTORC1-independent function. This study suggests that GATOR2 coordinates amino acid and growth factor signaling to regulate oligodendrocyte myelination.
Collapse
Affiliation(s)
- Zongyan Yu
- School of Life Science and Technology, Harbin Institute of Technology, Harbin 150000, People's Republic of China
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518005, People's Republic of China
- Department of Biology, School of Life Sciences, Brain Research Center, Southern University of Science and Technology, Shenzhen 518000, People's Republic of China
| | - Zhiwen Yang
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518005, People's Republic of China
- Department of Biology, School of Life Sciences, Brain Research Center, Southern University of Science and Technology, Shenzhen 518000, People's Republic of China
| | - Guoru Ren
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518005, People's Republic of China
- Department of Biology, School of Life Sciences, Brain Research Center, Southern University of Science and Technology, Shenzhen 518000, People's Republic of China
| | - Yingjie Wang
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518005, People's Republic of China
- Department of Biology, School of Life Sciences, Brain Research Center, Southern University of Science and Technology, Shenzhen 518000, People's Republic of China
| | - Xiang Luo
- School of Life Science and Technology, Harbin Institute of Technology, Harbin 150000, People's Republic of China
- Department of Biology, School of Life Sciences, Brain Research Center, Southern University of Science and Technology, Shenzhen 518000, People's Republic of China
| | - Feiyan Zhu
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518005, People's Republic of China
- Department of Biology, School of Life Sciences, Brain Research Center, Southern University of Science and Technology, Shenzhen 518000, People's Republic of China
| | - Shouyang Yu
- Neuroscience & Metabolism Research, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, People's Republic of China
| | - Lanlan Jia
- Neuroscience & Metabolism Research, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, People's Republic of China
| | - Mina Chen
- Neuroscience & Metabolism Research, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, People's Republic of China
| | - Paul F Worley
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Bo Xiao
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518005, People's Republic of China;
- Department of Biology, School of Life Sciences, Brain Research Center, Southern University of Science and Technology, Shenzhen 518000, People's Republic of China
| |
Collapse
|
12
|
Zheng Z, Chen J, Chopp M. Mechanisms of Plasticity Remodeling and Recovery. Stroke 2022. [DOI: 10.1016/b978-0-323-69424-7.00011-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
13
|
Longo F, Klann E. Reciprocal control of translation and transcription in autism spectrum disorder. EMBO Rep 2021; 22:e52110. [PMID: 33977633 PMCID: PMC8183409 DOI: 10.15252/embr.202052110] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 02/20/2021] [Accepted: 04/19/2021] [Indexed: 12/11/2022] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by deficits in social communication and the presence of restricted patterns of interest and repetitive behaviors. ASD is genetically heterogeneous and is believed to be caused by both inheritable and de novo gene variations. Studies have revealed an extremely complex genetic landscape of ASD, favoring the idea that mutations in different clusters of genes interfere with interconnected downstream signaling pathways and circuitry, resulting in aberrant behavior. In this review, we describe a select group of candidate genes that represent both syndromic and non-syndromic forms of ASD and encode proteins that are important in transcriptional and translational regulation. We focus on the interplay between dysregulated translation and transcription in ASD with the hypothesis that dysregulation of each synthetic process triggers a feedback loop to act on the other, which ultimately exacerbates ASD pathophysiology. Finally, we summarize findings from interdisciplinary studies that pave the way for the investigation of the cooperative impact of different genes and pathways underlying the development of ASD.
Collapse
Affiliation(s)
| | - Eric Klann
- Center for Neural ScienceNew York UniversityNew YorkNYUSA
| |
Collapse
|
14
|
Dillinger AE, Kuespert S, Froemel F, Tamm ER, Fuchshofer R. CCN2/CTGF promotor activity in the developing and adult mouse eye. Cell Tissue Res 2021; 384:625-641. [PMID: 33512643 PMCID: PMC8211604 DOI: 10.1007/s00441-020-03332-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 10/29/2020] [Indexed: 12/23/2022]
Abstract
CCN2/CTGF is a matricellular protein that is known to enhance transforming growth factor-β signaling and to induce a myofibroblast-like phenotype in a variety of cell types. Here, we investigated Ccn2/Ctgf promotor activity during development and in the adult mouse eye, using CTGFLacZ/+ mice in which the β-galactosidase reporter gene LacZ had been inserted into the open reading frame of Ccn2/Ctgf. Promotor activity was assessed by staining for β-galactosidase activity and by immunolabeling using antibodies against β-galactosidase. Co-immunostaining using antibodies against glutamine synthetase, glial fibrillary acidic protein, choline acetyltransferase, and CD31 was applied to identify specific cell types. Ccn2/Ctgf promotor activity was intense in neural crest-derived cells differentiating to corneal stroma and endothelium, and to the stroma of choroid, iris, ciliary body, and the trabecular meshwork during development. In the adult eye, a persistent and very strong promotor activity was present in the trabecular meshwork outflow pathways. In addition, endothelial cells of Schlemm’s canal, and of retinal and choroidal vessels, retinal astrocytes, Müller glia, and starburst amacrine cells were stained. Very strong promoter activity was seen in the astrocytes of the glial lamina at the optic nerve head. We conclude that CCN2/CTGF signaling is involved in the processes that govern neural crest morphogenesis during ocular development. In the adult eye, CCN2/CTGF likely plays an important role for the trabecular meshwork outflow pathways and the glial lamina of the optic nerve head.
Collapse
Affiliation(s)
- Andrea E Dillinger
- Institute of Human Anatomy and Embryology, University of Regensburg, 93053, Regensburg, Germany
| | - Sabrina Kuespert
- Institute of Human Anatomy and Embryology, University of Regensburg, 93053, Regensburg, Germany
| | - Franziska Froemel
- Institute of Human Anatomy and Embryology, University of Regensburg, 93053, Regensburg, Germany
| | - Ernst R Tamm
- Institute of Human Anatomy and Embryology, University of Regensburg, 93053, Regensburg, Germany
| | - Rudolf Fuchshofer
- Institute of Human Anatomy and Embryology, University of Regensburg, 93053, Regensburg, Germany.
| |
Collapse
|
15
|
SRF Is Required for Maintenance of Astrocytes in Non-Reactive State in the Mammalian Brain. eNeuro 2021; 8:ENEURO.0447-19.2020. [PMID: 33441399 PMCID: PMC7877468 DOI: 10.1523/eneuro.0447-19.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 12/24/2020] [Accepted: 12/24/2020] [Indexed: 11/21/2022] Open
Abstract
Astrocytes play several critical roles in the normal functioning of the mammalian brain, including ion homeostasis, synapse formation, and synaptic plasticity. Following injury and infection or in the setting of neurodegeneration, astrocytes become hypertrophic and reactive, a process termed astrogliosis. Although acute reactive gliosis is beneficial in limiting further tissue damage, chronic gliosis becomes detrimental for neuronal recovery and regeneration. Several extracellular factors have been identified that generate reactive astrocytes; however, very little is known about the cell-autonomous transcriptional mechanisms that regulate the maintenance of astrocytes in the normal non-reactive state. Here, we show that conditional deletion of the stimulus-dependent transcription factor, serum response factor (SRF) in astrocytes (SrfGFAPCKO) results in astrogliosis marked by hypertrophic morphology and increased expression of GFAP, vimentin, and nestin. These reactive astrocytes were not restricted to any specific brain region and were seen in both white and gray matter in the entire brain. This astrogliosis persisted throughout adulthood concomitant with microglial activation. Importantly, the Srf mutant mouse brain did not exhibit any cell death or blood brain barrier (BBB) deficits suggesting that apoptosis and leaky BBB are not the causes for the reactive phenotype. The mutant astrocytes expressed more A2 reactive astrocyte marker genes and the SrfGFAPCKO mice exhibited normal neuronal numbers indicating that SRF-deficient gliosis astrocytes are not neurotoxic. Together, our findings suggest that SRF plays a critical role in astrocytes to maintain them in a non-reactive state.
Collapse
|
16
|
Tsai SH, Tsao CY, Lee LJ. Altered White Matter and Layer VIb Neurons in Heterozygous Disc1 Mutant, a Mouse Model of Schizophrenia. Front Neuroanat 2020; 14:605029. [PMID: 33384588 PMCID: PMC7769951 DOI: 10.3389/fnana.2020.605029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 11/24/2020] [Indexed: 11/13/2022] Open
Abstract
Increased white matter neuron density has been associated with neuropsychiatric disorders including schizophrenia. However, the pathogenic features of these neurons are still largely unknown. Subplate neurons, the earliest generated neurons in the developing cortex have also been associated with schizophrenia and autism. The link between these neurons and mental disorders is also not well established. Since cortical layer VIb neurons are believed to be the remnant of subplate neurons in the adult rodent brain, in this study, we aimed to examine the cytoarchitecture of neurons in cortical layer VIb and the underlying white matter in heterozygous Disc1 mutant (Het) mice, a mouse model of schizophrenia. In the white matter, the number of NeuN-positive neurons was quite low in the external capsule; however, the density of these cells was found increased (54%) in Het mice compared with wildtype (WT) littermates. The density of PV-positive neurons was unchanged in the mutants. In the cortical layer VIb, the density of CTGF-positive neurons increased (21.5%) in Het mice, whereas the number of Cplx3-positive cells reduced (16.1%) in these mutants, compared with WT mice. Layer VIb neurons can be classified by their morphological characters. The morphology of Type I pyramidal neurons was comparable between genotypes while the dendritic length and complexity of Type II multipolar neurons were significantly reduced in Het mice. White matter neurons and layer VIb neurons receive synaptic inputs and modulate the process of sensory information and sleep/arousal pattern. Aberrances of these neurons in Disc1 mutants implies altered brain functions in these mice.
Collapse
Affiliation(s)
- Shin-Hwa Tsai
- School of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chih-Yu Tsao
- Graduate Institute of Anatomy and Cell Biology, National Taiwan University, Taipei, Taiwan
| | - Li-Jen Lee
- School of Medicine, National Taiwan University, Taipei, Taiwan
- Graduate Institute of Anatomy and Cell Biology, National Taiwan University, Taipei, Taiwan
- Institute of Brain and Mind Sciences, National Taiwan University, Taipei, Taiwan
- Neurobiology and Cognitive Science Center, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
17
|
Mice Lacking Connective Tissue Growth Factor in the Forebrain Exhibit Delayed Seizure Response, Reduced C-Fos Expression and Different Microglial Phenotype Following Acute PTZ Injection. Int J Mol Sci 2020; 21:ijms21144921. [PMID: 32664674 PMCID: PMC7404259 DOI: 10.3390/ijms21144921] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 06/29/2020] [Accepted: 07/10/2020] [Indexed: 01/03/2023] Open
Abstract
Connective tissue growth factor (CTGF) plays important roles in the development and regeneration of the connective tissue, yet its function in the nervous system is still not clear. CTGF is expressed in some distinct regions of the brain, including the dorsal endopiriform nucleus (DEPN) which has been recognized as an epileptogenic zone. We generated a forebrain-specific Ctgf knockout (FbCtgf KO) mouse line in which the expression of Ctgf in the DEPN is eliminated. In this study, we adopted a pentylenetetrazole (PTZ)-induced seizure model and found similar severity and latencies to death between FbCtgf KO and WT mice. Interestingly, there was a delay in the seizure reactions in the mutant mice. We further observed reduced c-fos expression subsequent to PTZ treatment in the KO mice, especially in the hippocampus. While the densities of astrocytes and microglia in the hippocampus were kept constant after acute PTZ treatment, microglial morphology was different between genotypes. Our present study demonstrated that in the FbCtgf KO mice, PTZ failed to increase neuronal activity and microglial response in the hippocampus. Our results suggested that inhibition of Ctgf function may have a therapeutic potential in preventing the pathophysiology of epilepsy.
Collapse
|
18
|
Dynamic CCN3 expression in the murine CNS does not confer essential roles in myelination or remyelination. Proc Natl Acad Sci U S A 2020; 117:18018-18028. [PMID: 32651278 PMCID: PMC7395501 DOI: 10.1073/pnas.1922089117] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Remyelination is a natural regenerative process driven by oligodendrocytes that occurs following myelin damage. Understanding this process holds therapeutic value for demyelinating diseases such as multiple sclerosis, in which remyelination can fail. CCN3 is a matricellular protein previously reported to enhance oligodendrocyte progenitor differentiation and myelination in vitro and ex vivo. Here, we show that despite extensive and dynamic expression in the murine CNS in homeostasis and following toxin-induced myelin damage, CCN3 is not required for myelination or remyelination in vivo. Yet, the anatomically distinct expression pattern suggests unidentified roles of CCN3 in a range of neurological processes. This investigation provides a framework for future investigations of the expression and role of CCN proteins in the CNS. CCN3 is a matricellular protein that promotes oligodendrocyte progenitor cell differentiation and myelination in vitro and ex vivo. CCN3 is therefore a candidate of interest in central nervous system (CNS) myelination and remyelination, and we sought to investigate the expression and role of CCN3 during these processes. We found CCN3 to be expressed predominantly by neurons in distinct areas of the CNS, primarily the cerebral cortex, hippocampus, amygdala, suprachiasmatic nuclei, anterior olfactory nuclei, and spinal cord gray matter. CCN3 was transiently up-regulated following demyelination in the brain of cuprizone-fed mice and spinal cord lesions of mice injected with lysolecithin. However, CCN3−/− mice did not exhibit significantly different numbers of oligodendroglia or differentiated oligodendrocytes in the healthy or remyelinating CNS, compared to WT controls. These results suggest that despite robust and dynamic expression in the CNS, CCN3 is not required for efficient myelination or remyelination in the murine CNS in vivo.
Collapse
|
19
|
Gerosa L, Grillo B, Forastieri C, Longaretti A, Toffolo E, Mallei A, Bassani S, Popoli M, Battaglioli E, Rusconi F. SRF and SRFΔ5 Splicing Isoform Recruit Corepressor LSD1/KDM1A Modifying Structural Neuroplasticity and Environmental Stress Response. Mol Neurobiol 2019; 57:393-407. [DOI: 10.1007/s12035-019-01720-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
20
|
Yu IS, Chang HC, Chen KC, Lu YL, Shy HT, Chen CY, Lee KY, Lee LJ. Genetic Elimination of Connective Tissue Growth Factor in the Forebrain Affects Subplate Neurons in the Cortex and Oligodendrocytes in the Underlying White Matter. Front Neuroanat 2019; 13:16. [PMID: 30842729 PMCID: PMC6391576 DOI: 10.3389/fnana.2019.00016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 02/01/2019] [Indexed: 12/23/2022] Open
Abstract
Connective tissue growth factor (CTGF) is a secreted extracellular matrix-associated protein, which play a role in regulating various cellular functions. Although the expression of CTGF has been reported in the cortical subplate, its function is still not clear. Thus, to explore the significance of CTGF in the brain, we created a forebrain-specific Ctgf knockout (FbCtgf KO) mouse model. By crossing Ctgffl/fl mice with Emx1-Cre transgenic mice, in which the expression of Cre is prenatally initiated, the full length Ctgf is removed in the forebrain structures. In young adult (2–3 months old) FbCtgf KO mice, subplate markers such as Nurr1 and Cplx3 are still expressed in the cortical layer VIb; however, the density of the subplate neurons is increased. Interestingly, in these mutants, we found a reduced structural complexity in the subplate neurons. The distribution patterns of neurons and glial cells, examined by immunohistochemistry, are comparable between genotypes in the somatosensory cortex. However, increased densities of mature oligodendrocytes, but not immature ones, were noticed in the external capsule underneath the cortical layer VIb in young adult FbCtgf KO mice. The features of myelinated axons in the external capsule were then examined using electron microscopy. Unexpectedly, the thickness of the myelin sheath was reduced in middle-aged (>12 months old), but not young adult FbCtgf KO mice. Our results suggest a secretory function of the subplate neurons, through the release of CTGF, which regulates the density and dendritic branching of subplate neurons as well as the maturation and function of nearby oligodendrocytes in the white matter.
Collapse
Affiliation(s)
- I-Shing Yu
- Laboratory Animal Center, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ho-Ching Chang
- Graduate Institute of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ko-Chien Chen
- Graduate Institute of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yi-Ling Lu
- Graduate Institute of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Horng-Tzer Shy
- Graduate Institute of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chwen-Yu Chen
- Department of Neurology, Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Kuang-Yung Lee
- Department of Neurology, Chang Gung Memorial Hospital, Keelung, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Li-Jen Lee
- Graduate Institute of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan.,Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei, Taiwan.,Neurobiology and Cognitive Science Center, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
21
|
Gonzalez D, Brandan E. CTGF/CCN2 from Skeletal Muscle to Nervous System: Impact on Neurodegenerative Diseases. Mol Neurobiol 2019; 56:5911-5916. [PMID: 30689195 DOI: 10.1007/s12035-019-1490-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 01/10/2019] [Indexed: 12/20/2022]
Abstract
Connective tissue growth factor (CTGF/CCN2) is a matricellular protein that belongs to the CCN family of proteins. Since its discovery, it has been linked to cellular processes such as cell proliferation, differentiation, adhesion, migration, and synthesis of extracellular matrix (ECM) components, among others. The pro-fibrotic role of CTGF/CCN2 has been well-studied in several pathologies characterized by the development of fibrosis. Reduction of CTGF/CCN2 levels in mdx mice, a murine model for Duchenne muscular dystrophy (DMD), decreases fibrosis and improves skeletal muscle phenotype and function. Recently, it has been shown that skeletal muscle of symptomatic hSOD1G93A mice, a model for Amyotrophic lateral sclerosis (ALS), shows up-regulation of CTGF/CCN2 accompanied by excessive deposition ECM molecules. Elevated levels of CTGF/CCN2 in spinal cord from ALS patients have been previously reported. However, there is no evidence regarding the role of CTGF/CCN2 in neurodegenerative diseases such as ALS, in which alterations in skeletal muscle seem to be the consequence of early pathological denervation. In this regard, the emerging evidence shows that CTGF/CCN2 also exerts non-fibrotic roles in the central nervous system (CNS), specifically impairing oligodendrocyte maturation and regeneration, and inhibiting axon myelination. Despite these striking observations, there is no evidence showing the role of CTGF/CCN2 in peripheral nerves. Therefore, even though more studies are needed to elucidate its precise role, CTGF/CCN2 is starting to emerge as a novel therapeutic target for the treatment of neurodegenerative diseases where demyelination and axonal degeneration occurs.
Collapse
Affiliation(s)
- David Gonzalez
- Centro de Envejecimiento y Regeneración, CARE Chile UC, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Libertador Bernardo O'Higgins 340, 8331150, Santiago, Chile
| | - Enrique Brandan
- Centro de Envejecimiento y Regeneración, CARE Chile UC, Pontificia Universidad Católica de Chile, Santiago, Chile. .,Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Libertador Bernardo O'Higgins 340, 8331150, Santiago, Chile.
| |
Collapse
|
22
|
de la Vega Gallardo N, Dittmer M, Dombrowski Y, Fitzgerald DC. Regenerating CNS myelin: Emerging roles of regulatory T cells and CCN proteins. Neurochem Int 2018; 130:104349. [PMID: 30513363 DOI: 10.1016/j.neuint.2018.11.024] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 11/19/2018] [Accepted: 11/30/2018] [Indexed: 02/08/2023]
Abstract
Efficient myelin regeneration in the central nervous system (CNS) requires the migration, proliferation and differentiation of oligodendrocyte progenitor cells (OPC) into myelinating oligodendrocytes. In demyelinating diseases such as multiple sclerosis (MS), this regenerative process can fail, and therapies targeting myelin repair are currently completely lacking in the clinic. The immune system is emerging as a key regenerative player in many tissues, such as muscle and heart. We recently reported that regulatory T cells (Treg) are required for efficient CNS remyelination. Furthermore, Treg secrete CCN3, a matricellular protein from the CCN family, implicated in regeneration of other tissues. Treg-derived CCN3 promoted oligodendrocyte differentiation and myelination. In contrast, previous studies showed that CCN2 inhibited myelination. These studies highlight the need for further scrutiny of the roles that CCN proteins play in myelin development and regeneration. Collectively, these findings open up exciting avenues of research to uncover the regenerative potential of the adaptive immune system.
Collapse
Affiliation(s)
- Nira de la Vega Gallardo
- Wellcome Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Northern Ireland, UK
| | - Marie Dittmer
- Wellcome Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Northern Ireland, UK
| | - Yvonne Dombrowski
- Wellcome Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Northern Ireland, UK
| | - Denise C Fitzgerald
- Wellcome Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Northern Ireland, UK.
| |
Collapse
|
23
|
Eagle AL, Gajewski PA, Robison AJ. Role of hippocampal activity-induced transcription in memory consolidation. Rev Neurosci 2018; 27:559-73. [PMID: 27180338 DOI: 10.1515/revneuro-2016-0010] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 03/26/2016] [Indexed: 01/15/2023]
Abstract
Experience-dependent changes in the strength of connections between neurons in the hippocampus (HPC) are critical for normal learning and memory consolidation, and disruption of this process drives a variety of neurological and psychiatric diseases. Proper HPC function relies upon discrete changes in gene expression driven by transcription factors (TFs) induced by neuronal activity. Here, we describe the induction and function of many of the most well-studied HPC TFs, including cyclic-AMP response element binding protein, serum-response factor, AP-1, and others, and describe their role in the learning process. We also discuss the known target genes of many of these TFs and the purported mechanisms by which they regulate long-term changes in HPC synaptic strength. Moreover, we propose that future research in this field will depend upon unbiased identification of additional gene targets for these activity-dependent TFs and subsequent meta-analyses that identify common genes or pathways regulated by multiple TFs in the HPC during learning or disease.
Collapse
|
24
|
Associating transcription factors and conserved RNA structures with gene regulation in the human brain. Sci Rep 2017; 7:5776. [PMID: 28720872 PMCID: PMC5516038 DOI: 10.1038/s41598-017-06200-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 06/20/2017] [Indexed: 02/06/2023] Open
Abstract
Anatomical subdivisions of the human brain can be associated with different neuronal functions. This functional diversification is reflected by differences in gene expression. By analyzing post-mortem gene expression data from the Allen Brain Atlas, we investigated the impact of transcription factors (TF) and RNA secondary structures on the regulation of gene expression in the human brain. First, we modeled the expression of a gene as a linear combination of the expression of TFs. We devised an approach to select robust TF-gene interactions and to determine localized contributions to gene expression of TFs. Among the TFs with the most localized contributions, we identified EZH2 in the cerebellum, NR3C1 in the cerebral cortex and SRF in the basal forebrain. Our results suggest that EZH2 is involved in regulating ZIC2 and SHANK1 which have been linked to neurological diseases such as autism spectrum disorder. Second, we associated enriched regulatory elements inside differentially expressed mRNAs with RNA secondary structure motifs. We found a group of purine-uracil repeat RNA secondary structure motifs plus other motifs in neuron related genes such as ACSL4 and ERLIN2.
Collapse
|
25
|
Millar LJ, Shi L, Hoerder-Suabedissen A, Molnár Z. Neonatal Hypoxia Ischaemia: Mechanisms, Models, and Therapeutic Challenges. Front Cell Neurosci 2017; 11:78. [PMID: 28533743 PMCID: PMC5420571 DOI: 10.3389/fncel.2017.00078] [Citation(s) in RCA: 231] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 03/07/2017] [Indexed: 12/11/2022] Open
Abstract
Neonatal hypoxia-ischaemia (HI) is the most common cause of death and disability in human neonates, and is often associated with persistent motor, sensory, and cognitive impairment. Improved intensive care technology has increased survival without preventing neurological disorder, increasing morbidity throughout the adult population. Early preventative or neuroprotective interventions have the potential to rescue brain development in neonates, yet only one therapeutic intervention is currently licensed for use in developed countries. Recent investigations of the transient cortical layer known as subplate, especially regarding subplate's secretory role, opens up a novel set of potential molecular modulators of neonatal HI injury. This review examines the biological mechanisms of human neonatal HI, discusses evidence for the relevance of subplate-secreted molecules to this condition, and evaluates available animal models. Neuroserpin, a neuronally released neuroprotective factor, is discussed as a case study for developing new potential pharmacological interventions for use post-ischaemic injury.
Collapse
Affiliation(s)
- Lancelot J. Millar
- Molnár Group, Department of Physiology, Anatomy and Genetics, University of OxfordOxford, UK
| | - Lei Shi
- Molnár Group, Department of Physiology, Anatomy and Genetics, University of OxfordOxford, UK
- JNU-HKUST Joint Laboratory for Neuroscience and Innovative Drug Research, College of Pharmacy, Jinan UniversityGuangzhou, China
| | | | - Zoltán Molnár
- Molnár Group, Department of Physiology, Anatomy and Genetics, University of OxfordOxford, UK
| |
Collapse
|
26
|
Pfisterer U, Khodosevich K. Neuronal survival in the brain: neuron type-specific mechanisms. Cell Death Dis 2017; 8:e2643. [PMID: 28252642 PMCID: PMC5386560 DOI: 10.1038/cddis.2017.64] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 01/24/2017] [Accepted: 01/31/2017] [Indexed: 12/19/2022]
Abstract
Neurogenic regions of mammalian brain produce many more neurons that will eventually survive and reach a mature stage. Developmental cell death affects both embryonically produced immature neurons and those immature neurons that are generated in regions of adult neurogenesis. Removal of substantial numbers of neurons that are not yet completely integrated into the local circuits helps to ensure that maturation and homeostatic function of neuronal networks in the brain proceed correctly. External signals from brain microenvironment together with intrinsic signaling pathways determine whether a particular neuron will die. To accommodate this signaling, immature neurons in the brain express a number of transmembrane factors as well as intracellular signaling molecules that will regulate the cell survival/death decision, and many of these factors cease being expressed upon neuronal maturation. Furthermore, pro-survival factors and intracellular responses depend on the type of neuron and region of the brain. Thus, in addition to some common neuronal pro-survival signaling, different types of neurons possess a variety of 'neuron type-specific' pro-survival constituents that might help them to adapt for survival in a certain brain region. This review focuses on how immature neurons survive during normal and impaired brain development, both in the embryonic/neonatal brain and in brain regions associated with adult neurogenesis, and emphasizes neuron type-specific mechanisms that help to survive for various types of immature neurons. Importantly, we mainly focus on in vivo data to describe neuronal survival specifically in the brain, without extrapolating data obtained in the PNS or spinal cord, and thus emphasize the influence of the complex brain environment on neuronal survival during development.
Collapse
Affiliation(s)
- Ulrich Pfisterer
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Konstantin Khodosevich
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
27
|
Ercan E, Han JM, Di Nardo A, Winden K, Han MJ, Hoyo L, Saffari A, Leask A, Geschwind DH, Sahin M. Neuronal CTGF/CCN2 negatively regulates myelination in a mouse model of tuberous sclerosis complex. J Exp Med 2017; 214:681-697. [PMID: 28183733 PMCID: PMC5339668 DOI: 10.1084/jem.20160446] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 11/09/2016] [Accepted: 12/30/2016] [Indexed: 12/11/2022] Open
Abstract
Disruption of myelination during development has been implicated in a range of neurodevelopmental disorders including tuberous sclerosis complex (TSC). TSC patients with autism display impairments in white matter integrity. Similarly, mice lacking neuronal Tsc1 have a hypomyelination phenotype. However, the mechanisms that underlie these phenotypes remain unknown. In this study, we demonstrate that neuronal TSC1/2 orchestrates a program of oligodendrocyte maturation through the regulated secretion of connective tissue growth factor (CTGF). We characterize oligodendrocyte maturation both in vitro and in vivo. We find that neuron-specific Tsc1 deletion results in an increase in CTGF secretion that non-cell autonomously stunts oligodendrocyte development and decreases the total number of oligodendrocytes. Genetic deletion of CTGF from neurons, in turn, mitigates the TSC-dependent hypomyelination phenotype. These results show that the mechanistic target of rapamycin (mTOR) pathway in neurons regulates CTGF production and secretion, revealing a paracrine mechanism by which neuronal signaling regulates oligodendrocyte maturation and myelination in TSC. This study highlights the role of mTOR-dependent signaling between neuronal and nonneuronal cells in the regulation of myelin and identifies an additional therapeutic avenue for this disease.
Collapse
Affiliation(s)
- Ebru Ercan
- Department of Neurology, F.M. Kirby Center for Neurobiology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115
| | - Juliette M Han
- Department of Neurology, F.M. Kirby Center for Neurobiology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115
| | - Alessia Di Nardo
- Department of Neurology, F.M. Kirby Center for Neurobiology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115
| | - Kellen Winden
- Department of Neurology, F.M. Kirby Center for Neurobiology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115
| | - Min-Joon Han
- Department of Neurology, F.M. Kirby Center for Neurobiology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115.,Translational Neuroscience Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115
| | - Leonie Hoyo
- Department of Neurology, F.M. Kirby Center for Neurobiology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115.,Translational Neuroscience Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115
| | - Afshin Saffari
- Department of Neurology, F.M. Kirby Center for Neurobiology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115
| | - Andrew Leask
- Department of Dentistry, Schulich School of Medicine and Dentistry, London, Ontario N6A 5C1, Canada.,Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, London, Ontario N6A 5C1, Canada
| | - Daniel H Geschwind
- Department of Neurology, University of California, Los Angeles School of Medicine, Los Angeles, CA 90095.,Semel Institute, University of California, Los Angeles School of Medicine, Los Angeles, CA 90095.,Department of Human Genetics, University of California, Los Angeles School of Medicine, Los Angeles, CA 90095
| | - Mustafa Sahin
- Department of Neurology, F.M. Kirby Center for Neurobiology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115 .,Translational Neuroscience Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115
| |
Collapse
|
28
|
Serum Response Factor (SRF) Ablation Interferes with Acute Stress-Associated Immediate and Long-Term Coping Mechanisms. Mol Neurobiol 2016; 54:8242-8262. [PMID: 27914009 DOI: 10.1007/s12035-016-0300-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 11/14/2016] [Indexed: 12/26/2022]
Abstract
Stress experience modulates behavior, metabolism, and energy expenditure of organisms. One molecular hallmark of an acute stress response is a rapid induction of immediate early genes (IEGs) such as c-Fos and Egr family members. IEG transcription in neurons is mediated by the neuronal activity-driven gene regulator serum response factor (SRF). We show a first role of SRF in immediate and long-lasting acute restraint stress (AS) responses. For this, we employed a standardized mouse phenotyping protocol at the German Mouse Clinic (GMC) including behavioral, metabolic, and cardiologic tests as well as gene expression profiling to analyze the consequences of forebrain-specific SRF deletion in mice exposed to AS. Adult mice with an SRF deletion in glutamatergic neurons (Srf; CaMKIIa-CreERT2 ) showed hyperactivity, decreased anxiety, and impaired working memory. In response to restraint AS, instant stress reactivity including locomotor behavior and corticosterone induction was impaired in Srf mutant mice. Interestingly, even several weeks after previous AS exposure, SRF-deficient mice showed long-lasting AS-associated changes including altered locomotion, metabolism, energy expenditure, and cardiovascular changes. This suggests a requirement of SRF for mediating long-term stress coping mechanisms in wild-type mice. SRF ablation decreased AS-mediated IEG induction and activity of the actin severing protein cofilin. In summary, our data suggest an SRF function in immediate AS reactions and long-term post-stress-associated coping mechanisms.
Collapse
|
29
|
Muehlich S, Hermanns C, Meier MA, Kircher P, Gudermann T. Unravelling a new mechanism linking actin polymerization and gene transcription. Nucleus 2016; 7:121-5. [PMID: 27104924 DOI: 10.1080/19491034.2016.1171433] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
In the recent years, the role of actin and actin-binding proteins in gene transcription has received considerable attention. Nuclear monomeric and polymerized actin and several actin binding proteins have been detected in the mammalian cell nucleus, although their roles in transcription are just beginning to emerge. Our group recently reported that the actin-binding protein Filamin A interacts with the transcriptional coactivator MKL1 to link actin polymerization with transcriptional activity of Serum Response Factor. Here we summarize the regulation and function of MKL1, and highlight this novel mechanism of MKL1 regulation through binding to Filamin A and its implications for cell migration.
Collapse
Affiliation(s)
- Susanne Muehlich
- a Walther Straub Institute of Pharmacology and Toxicology , Ludwig-Maximilians-University , Munich , Germany
| | - Constanze Hermanns
- a Walther Straub Institute of Pharmacology and Toxicology , Ludwig-Maximilians-University , Munich , Germany
| | - Melanie A Meier
- a Walther Straub Institute of Pharmacology and Toxicology , Ludwig-Maximilians-University , Munich , Germany
| | - Philipp Kircher
- a Walther Straub Institute of Pharmacology and Toxicology , Ludwig-Maximilians-University , Munich , Germany
| | - Thomas Gudermann
- a Walther Straub Institute of Pharmacology and Toxicology , Ludwig-Maximilians-University , Munich , Germany
| |
Collapse
|
30
|
Kuzniewska B, Nader K, Dabrowski M, Kaczmarek L, Kalita K. Adult Deletion of SRF Increases Epileptogenesis and Decreases Activity-Induced Gene Expression. Mol Neurobiol 2016; 53:1478-1493. [PMID: 25636686 PMCID: PMC4789231 DOI: 10.1007/s12035-014-9089-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 12/29/2014] [Indexed: 11/27/2022]
Abstract
Although the transcription factor serum response factor (SRF) has been suggested to play a role in activity-dependent gene expression and mediate plasticity-associated structural changes in the hippocampus, no unequivocal evidence has been provided for its role in brain pathology, such as epilepsy. A genome-wide program of activity-induced genes that are regulated by SRF also remains unknown. In the present study, we show that the inducible and conditional deletion of SRF in the adult mouse hippocampus increases the epileptic phenotype in the kainic acid model of epilepsy, reflected by more severe and frequent seizures. Moreover, we observe a robust decrease in activity-induced gene transcription in SRF knockout mice. We characterize the genetic program controlled by SRF in neurons and using functional annotation, we find that SRF target genes are associated with synaptic plasticity and epilepsy. Several of these SRF targets function as regulators of inhibitory or excitatory balance and the structural plasticity of neurons. Interestingly, mutations in those SRF targets have found to be associated with such human neuropsychiatric disorders, as autism and intellectual disability. We also identify novel direct SRF targets in hippocampus: Npas4, Gadd45g, and Zfp36. Altogether, our data indicate that proteins that are highly upregulated by neuronal stimulation, identified in the present study as SRF targets, may function as endogenous protectors against overactivation. Thus, the lack of these effector proteins in SRF knockout animals may lead to uncontrolled excitation and eventually epilepsy.
Collapse
Affiliation(s)
- Bozena Kuzniewska
- Laboratory of Neurobiology, Nencki Institute, 3 Pasteur Street, Warsaw, Poland
| | - Karolina Nader
- Laboratory of Neurobiology, Nencki Institute, 3 Pasteur Street, Warsaw, Poland
| | - Michal Dabrowski
- Laboratory of Bioinformatics, Neurobiology Center, Nencki Institute, 3 Pasteur Street, Warsaw, Poland
| | - Leszek Kaczmarek
- Laboratory of Neurobiology, Nencki Institute, 3 Pasteur Street, Warsaw, Poland
| | - Katarzyna Kalita
- Laboratory of Neurobiology, Nencki Institute, 3 Pasteur Street, Warsaw, Poland.
| |
Collapse
|
31
|
LSD1 modulates stress-evoked transcription of immediate early genes and emotional behavior. Proc Natl Acad Sci U S A 2016; 113:3651-6. [PMID: 26976584 DOI: 10.1073/pnas.1511974113] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Behavioral changes in response to stressful stimuli can be controlled via adaptive epigenetic changes in neuronal gene expression. Here we indicate a role for the transcriptional corepressor Lysine-Specific Demethylase 1 (LSD1) and its dominant-negative splicing isoform neuroLSD1, in the modulation of emotional behavior. In mouse hippocampus, we show that LSD1 and neuroLSD1 can interact with transcription factor serum response factor (SRF) and set the chromatin state of SRF-targeted genes early growth response 1 (egr1) and c-fos Deletion or reduction of neuro LSD1 in mutant mice translates into decreased levels of activating histone marks at egr1 and c-fos promoters, dampening their psychosocial stress-induced transcription and resulting in low anxiety-like behavior. Administration of suberoylanilide hydroxamine to neuroLSD1(KO)mice reactivates egr1 and c-fos transcription and restores the behavioral phenotype. These findings indicate that LSD1 is a molecular transducer of stressful stimuli as well as a stress-response modifier. Indeed, LSD1 expression itself is increased acutely at both the transcriptional and splicing levels by psychosocial stress, suggesting that LSD1 is involved in the adaptive response to stress.
Collapse
|
32
|
Buffet C, Catelli MG, Hecale-Perlemoine K, Bricaire L, Garcia C, Gallet-Dierick A, Rodriguez S, Cormier F, Groussin L. Dual Specificity Phosphatase 5, a Specific Negative Regulator of ERK Signaling, Is Induced by Serum Response Factor and Elk-1 Transcription Factor. PLoS One 2015; 10:e0145484. [PMID: 26691724 PMCID: PMC4687125 DOI: 10.1371/journal.pone.0145484] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Accepted: 10/29/2015] [Indexed: 12/11/2022] Open
Abstract
Serum stimulation of mammalian cells induces, via the MAPK pathway, the nuclear protein DUSP5 (dual-specificity phosphatase 5), which specifically interacts with and inactivates the ERK1/2 MAP kinases. However, molecular mechanisms underlying DUSP5 induction are not well known. Here, we found that the DUSP5 mRNA induction depends on a transcriptional regulation by the MAPK pathway, without any modification of the mRNA stability. Two contiguous CArG boxes that bind serum response factor (SRF) were found in a 1 Kb promoter region, as well as several E twenty-six transcription factor family binding sites (EBS). These sites potentially bind Elk-1, a transcription factor activated by ERK1/2. Using wild type or mutated DUSP5 promoter reporters, we demonstrated that SRF plays a crucial role in serum induction of DUSP5 promoter activity, the proximal CArG box being important for SRF binding in vitro and in living cells. Moreover, in vitro and in vivo binding data of Elk-1 to the same promoter region further demonstrate a role for Elk-1 in the transcriptional regulation of DUSP5. SRF and Elk-1 form a ternary complex (Elk-1-SRF-DNA) on DUSP5 promoter, consequently providing a link to an important negative feedback tightly regulating phosphorylated ERK levels.
Collapse
Affiliation(s)
- Camille Buffet
- Endocrinology-Metabolism-Diabetes Department, Institut Cochin, Université Paris Descartes, CNRS (UMR8104), INSERM U1016, Paris, France
| | - Maria-Grazia Catelli
- Endocrinology-Metabolism-Diabetes Department, Institut Cochin, Université Paris Descartes, CNRS (UMR8104), INSERM U1016, Paris, France
| | - Karine Hecale-Perlemoine
- Endocrinology-Metabolism-Diabetes Department, Institut Cochin, Université Paris Descartes, CNRS (UMR8104), INSERM U1016, Paris, France
| | - Léopoldine Bricaire
- Endocrinology-Metabolism-Diabetes Department, Institut Cochin, Université Paris Descartes, CNRS (UMR8104), INSERM U1016, Paris, France
| | - Camille Garcia
- Endocrinology-Metabolism-Diabetes Department, Institut Cochin, Université Paris Descartes, CNRS (UMR8104), INSERM U1016, Paris, France
| | - Anne Gallet-Dierick
- Endocrinology-Metabolism-Diabetes Department, Institut Cochin, Université Paris Descartes, CNRS (UMR8104), INSERM U1016, Paris, France
| | - Stéphanie Rodriguez
- Endocrinology-Metabolism-Diabetes Department, Institut Cochin, Université Paris Descartes, CNRS (UMR8104), INSERM U1016, Paris, France
| | - Françoise Cormier
- Endocrinology-Metabolism-Diabetes Department, Institut Cochin, Université Paris Descartes, CNRS (UMR8104), INSERM U1016, Paris, France
| | - Lionel Groussin
- Endocrinology-Metabolism-Diabetes Department, Institut Cochin, Université Paris Descartes, CNRS (UMR8104), INSERM U1016, Paris, France
- Department of Endocrinology, Cochin Hospital, Paris, France
- * E-mail:
| |
Collapse
|
33
|
Angelini A, Li Z, Mericskay M, Decaux JF. Regulation of Connective Tissue Growth Factor and Cardiac Fibrosis by an SRF/MicroRNA-133a Axis. PLoS One 2015; 10:e0139858. [PMID: 26440278 PMCID: PMC4595333 DOI: 10.1371/journal.pone.0139858] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 09/16/2015] [Indexed: 01/26/2023] Open
Abstract
Myocardial fibrosis contributes to the remodeling of heart and the loss of cardiac function leading to heart failure. SRF is a transcription factor implicated in the regulation of a large variety of genes involved in cardiac structure and function. To investigate the impact of an SRF overexpression in heart, we developed a new cardiac-specific and tamoxifen-inducible SRF overexpression mouse model by the Cre/loxP strategy. Here, we report that a high level overexpression of SRF leads to severe modifications of cardiac cytoarchitecture affecting the balance between cardiomyocytes and cardiac fibroblasts and also a profound alteration of cardiac gene expression program. The drastic development of fibrosis was characterized by intense sirius red staining and associated with an increased expression of genes encoding extracellular matrix proteins such as fibronectin, procollagen type 1α1 and type 3α1 and especially connective tissue growth factor (CTGF). Furthermore miR-133a, one of the most predominant cardiac miRNAs, is strongly downregulated when SRF is overexpressed. By comparison a low level overexpression of SRF has minor impact on these different processes. Investigation with miR-133a, antimiR-133a and AdSRF-VP16 experiments in H9c2 cardiac cells demonstrated that: 1)–miR-133a acts as a repressor of SRF and CTGF expression; 2)–a simultaneous overexpression of SRF by AdSRF-VP16 and inhibition of miR-133a by a specific antimiR increase CTGF expression; 3)–miR-133a overexpression can block the upregulation of CTGF induced by AdSRF-VP16. Taken together, these findings reveal a key role of the SRF/CTGF/miR-133a axis in the regulation of cardiac fibrosis.
Collapse
Affiliation(s)
- Aude Angelini
- Biology of Adaptation and Ageing, Institut de Biologie Paris Seine (IBPS), DHU FAST Sorbonne Universités, UPMC Université Paris 06, Paris, France
- CNRS, UMR8256, Paris, France
- INSERM, U1164, Paris, France
| | - Zhenlin Li
- Biology of Adaptation and Ageing, Institut de Biologie Paris Seine (IBPS), DHU FAST Sorbonne Universités, UPMC Université Paris 06, Paris, France
- CNRS, UMR8256, Paris, France
- INSERM, U1164, Paris, France
| | - Mathias Mericskay
- Biology of Adaptation and Ageing, Institut de Biologie Paris Seine (IBPS), DHU FAST Sorbonne Universités, UPMC Université Paris 06, Paris, France
- CNRS, UMR8256, Paris, France
- INSERM, U1164, Paris, France
- * E-mail: (JD); (MM)
| | - Jean-François Decaux
- Biology of Adaptation and Ageing, Institut de Biologie Paris Seine (IBPS), DHU FAST Sorbonne Universités, UPMC Université Paris 06, Paris, France
- CNRS, UMR8256, Paris, France
- INSERM, U1164, Paris, France
- * E-mail: (JD); (MM)
| |
Collapse
|
34
|
Foxworthy WA, Medina AE. Overexpression of Serum Response Factor in Neurons Restores Ocular Dominance Plasticity in a Model of Fetal Alcohol Spectrum Disorders. Alcohol Clin Exp Res 2015; 39:1951-6. [PMID: 26342644 DOI: 10.1111/acer.12844] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 07/14/2015] [Indexed: 01/06/2023]
Abstract
BACKGROUND Deficits in neuronal plasticity underlie many neurobehavioral and cognitive problems presented in fetal alcohol spectrum disorder (FASD). Our laboratory has developed a ferret model showing that early alcohol exposure leads to a persistent disruption in ocular dominance plasticity (ODP). For instance, a few days of monocular deprivation results in a robust reduction of visual cortex neurons' responsiveness to stimulation of the deprived eye in normal animals, but not in ferrets with early alcohol exposure. Previously our laboratory demonstrated that overexpression of serum response factor (SRF) exclusively in astrocytes can improve neuronal plasticity in FASD. Here, we test whether neuronal overexpression of SRF can achieve similar effects. METHODS Ferrets received 3.5 g/kg alcohol intraperitoneally (25% in saline) or saline as control every other day between postnatal day 10 to 30, which is roughly equivalent to the third trimester of human gestation. Animals were given intracortical injections of a Herpes Simplex Virus-based vector to express either green fluorescent protein or a constitutively active form of SRF in infected neurons. They were then monocularly deprived by eyelid suture for 4 to 5 days after which single-unit recordings were conducted to determine whether changes in ocular dominance had occurred. RESULTS Overexpression of a constitutively active form of SRF by neurons restored ODP in alcohol-treated animals. This effect was observed only in areas near the site of viral infection. CONCLUSIONS Overexpression of SRF in neurons can restore plasticity in the ferret model of FASD, but only in areas near the site of infection. This contrasts with SRF overexpression in astrocytes which restored plasticity throughout the visual cortex.
Collapse
Affiliation(s)
- W Alex Foxworthy
- Department of Pediatrics (WAF, AEM), University of Maryland, Baltimore, Maryland
| | - Alexandre E Medina
- Department of Pediatrics (WAF, AEM), University of Maryland, Baltimore, Maryland
| |
Collapse
|
35
|
Two functional promoter polymorphisms of neuregulin 1 gene are associated with progressive forms of multiple sclerosis. J Neurol Sci 2015; 351:154-159. [DOI: 10.1016/j.jns.2015.03.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Revised: 02/06/2015] [Accepted: 03/04/2015] [Indexed: 01/19/2023]
|
36
|
Kondo S, Al-Hasani H, Hoerder-Suabedissen A, Wang WZ, Molnár Z. Secretory function in subplate neurons during cortical development. Front Neurosci 2015; 9:100. [PMID: 25859180 PMCID: PMC4374456 DOI: 10.3389/fnins.2015.00100] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 03/10/2015] [Indexed: 01/15/2023] Open
Abstract
Subplate cells are among the first generated neurons in the mammalian cerebral cortex and have been implicated in the establishment of cortical wiring. In rodents some subplate neurons persist into adulthood. Here we would like to highlight several converging findings which suggest a novel secretory function of subplate neurons during cortical development. Throughout the postnatal period in rodents, subplate neurons have highly developed rough endoplasmic reticulum (ER) and are under an ER stress condition. By comparing gene expression between subplate and layer 6, we found that several genes encoding secreted proteins are highly expressed in subplate neurons. One of these secreted proteins, neuroserpin, encoded by the serpini1 gene, is localized to the ER in subplate cells. We propose that subplate might influence cortical circuit formation through a transient secretory function.
Collapse
Affiliation(s)
- Shinichi Kondo
- Department of Physiology, Anatomy and Genetics, University of Oxford Oxford, UK
| | - Hannah Al-Hasani
- Department of Physiology, Anatomy and Genetics, University of Oxford Oxford, UK
| | | | - Wei Zhi Wang
- Department of Physiology, Anatomy and Genetics, University of Oxford Oxford, UK
| | - Zoltán Molnár
- Department of Physiology, Anatomy and Genetics, University of Oxford Oxford, UK
| |
Collapse
|
37
|
Shi N, Chen SY. From nerve to blood vessel: a new role of Olfm2 in smooth muscle differentiation from human embryonic stem cell-derived mesenchymal cells. J Biomed Res 2015; 29:261-3. [PMID: 26243513 PMCID: PMC4547375 DOI: 10.7555/jbr.29.20150027] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 03/08/2015] [Indexed: 01/10/2023] Open
Affiliation(s)
- Ning Shi
- Department of Physiology & Pharmacology, University of Georgia, Athens, GA 30602, USA
| | - Shi-You Chen
- Department of Physiology & Pharmacology, University of Georgia, Athens, GA 30602, USA.
| |
Collapse
|
38
|
Anastasiadou S, Liebenehm S, Sinske D, Meyer zu Reckendorf C, Moepps B, Nordheim A, Knöll B. Neuronal expression of the transcription factor serum response factor modulates myelination in a mouse multiple sclerosis model. Glia 2015; 63:958-76. [PMID: 25639799 DOI: 10.1002/glia.22794] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 01/08/2015] [Accepted: 01/08/2015] [Indexed: 12/14/2022]
Abstract
In multiple sclerosis (MS), neurons in addition to inflammatory cells are now considered to mediate disease origin and progression. So far, molecular and cellular mechanisms of neuronal MS contributions are poorly understood. Herein we analyzed whether neuron-restricted signaling by the neuroprotective transcription factor serum response factor (SRF) modulates de- and remyelination in a rodent MS model. In the mouse cuprizone model, neuron- (Srf (flox/flox;CaMKCreERT2)) but not glia-specific (Srf (flox/flox;PlpCreERT2)) SRF depletion impaired demyelination suggesting impaired debris clearance by astrocytes and microglia. This supports an important role of SRF expression in neurons but not oligodendrocytes in de- and remyelination. During remyelination, NG2- and OLIG2-positive cells of the oligodendrocyte lineage as well as de novo mRNA synthesis of myelin genes were also reduced in neuron-specific Srf mutants. Using the stripe assay, we demonstrate that cortices of cuprizone-fed wild-type mice elicited astrocyte and microglia activation whereas this was abrogated in cuprizone-fed neuron-specific Srf mutants. We identified CCL chemokines (e.g. CCL2) as neuron-derived SRF-regulated paracrine signals rescuing immune cell activation upon neuronal SRF deletion. In summary, we uncovered important roles of neurons and neuronally expressed SRF in MS associated de- and remyelination.
Collapse
Affiliation(s)
- Sofia Anastasiadou
- Institute of Physiological Chemistry, Ulm University, Albert-Einstein-Allee 11, 89081, Ulm, Germany
| | | | | | | | | | | | | |
Collapse
|
39
|
Anastasiadou S, Knöll B. Neuronal gene transcription modulates demyelination and remyelination in a mouse model of multiple sclerosis. Neural Regen Res 2015; 10:1401-2. [PMID: 26604895 PMCID: PMC4625500 DOI: 10.4103/1673-5374.165507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
|
40
|
Stern S, Knöll B. CNS axon regeneration inhibitors stimulate an immediate early gene response via MAP kinase-SRF signaling. Mol Brain 2014; 7:86. [PMID: 25406759 PMCID: PMC4243276 DOI: 10.1186/s13041-014-0086-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 11/11/2014] [Indexed: 12/24/2022] Open
Abstract
Background CNS axon regeneration inhibitors such as Nogo and CSPGs (Chondroitin Sulfate Proteoglycans) are major extrinsic factors limiting outgrowth of severed nerve fibers. However, knowledge on intracellular signaling cascades and gene expression programs activated by these inhibitors in neurons is sparse. Herein we studied intracellular signaling cascades activated by total myelin, Nogo and CSPGs in primary mouse CNS neurons. Results Total myelin, Nogo and CSPGs stimulated gene expression activity of the serum response factor (SRF), a central gene regulator of immediate early (IEG) and actin cytoskeletal gene transcription. As demonstrated by pharmacological interference, SRF-mediated IEG activation by myelin, Nogo or CSPGs depended on MAP kinase, to a lesser extent on Rho-GTPase but not on PKA signaling. Stimulation of neurons with all three axon growth inhibitors activated the MAP kinase ERK. In addition to ERK activation, myelin activated the IEG c-Fos, an important checkpoint of neuronal survival vs. apoptosis. Employing Srf deficient neurons revealed that myelin-induced IEG activation requires SRF. This suggests an SRF function in mediating neuronal signaling evoked by axon regeneration associated inhibitors. Besides being a signaling target of axon growth inhibitors, we show that constitutively-active SRF-VP16 can be employed to circumvent neurite growth inhibition imposed by myelin, Nogo and CSPGs. Conclusion In sum, our data demonstrate that axon regeneration inhibitors such as Nogo trigger gene expression programs including an SRF-dependent IEG response via MAP kinases and Rho-GTPases.
Collapse
Affiliation(s)
- Sina Stern
- Department Molecular Biology, Eberhard-Karls-University Tübingen, Interfaculty Institute for Cell Biology, Auf der Morgenstelle 15, 72076, Tübingen, Germany. .,Current address: German Centre for Neurodegenerative Diseases (DZNE), Ludwig-Erhard-Allee 2, 53175, Bonn, Germany.
| | - Bernd Knöll
- Department Molecular Biology, Eberhard-Karls-University Tübingen, Interfaculty Institute for Cell Biology, Auf der Morgenstelle 15, 72076, Tübingen, Germany. .,Current address: Ulm University, Institute for Physiological Chemistry, 89081, Ulm, Germany.
| |
Collapse
|
41
|
Vasudevan HN, Soriano P. SRF regulates craniofacial development through selective recruitment of MRTF cofactors by PDGF signaling. Dev Cell 2014; 31:332-344. [PMID: 25453829 DOI: 10.1016/j.devcel.2014.10.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Revised: 09/02/2014] [Accepted: 10/03/2014] [Indexed: 02/07/2023]
Abstract
Receptor tyrosine kinase signaling is critical for mammalian craniofacial development, but the key downstream transcriptional effectors remain unknown. We demonstrate that serum response factor (SRF) is induced by both platelet-derived growth factor (PDGF) and fibroblast growth factor (FGF) signaling in mouse embryonic palatal mesenchyme cells and that Srf neural crest conditional mutants exhibit facial clefting accompanied by proliferation and migration defects. Srf and Pdgfra mutants interact genetically in craniofacial development, but Srf and Fgfr1 mutants do not. This signal specificity is recapitulated at the level of cofactor activation: while both PDGF and FGF target gene promoters show enriched genome-wide overlap with SRF ChIP-seq peaks, PDGF selectively activates a network of MRTF-dependent cytoskeletal genes. Collectively, our results identify a role for SRF in proliferation and migration during craniofacial development and delineate a mechanism of receptor tyrosine kinase specificity mediated through differential cofactor usage, leading to a PDGF-responsive SRF-driven transcriptional program in the midface.
Collapse
Affiliation(s)
- Harish N Vasudevan
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Philippe Soriano
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
42
|
Brennan-Laun SE, Li XL, Ezelle HJ, Venkataraman T, Blackshear PJ, Wilson GM, Hassel BA. RNase L attenuates mitogen-stimulated gene expression via transcriptional and post-transcriptional mechanisms to limit the proliferative response. J Biol Chem 2014; 289:33629-43. [PMID: 25301952 DOI: 10.1074/jbc.m114.589556] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The cellular response to mitogens is tightly regulated via transcriptional and post-transcriptional mechanisms to rapidly induce genes that promote proliferation and efficiently attenuate their expression to prevent malignant growth. RNase L is an endoribonuclease that mediates diverse antiproliferative activities, and tristetraprolin (TTP) is a mitogen-induced RNA-binding protein that directs the decay of proliferation-stimulatory mRNAs. In light of their roles as endogenous proliferative constraints, we examined the mechanisms and functional interactions of RNase L and TTP to attenuate a mitogenic response. Mitogen stimulation of RNase L-deficient cells significantly increased TTP transcription and the induction of other mitogen-induced mRNAs. This regulation corresponded with elevated expression of serum-response factor (SRF), a master regulator of mitogen-induced transcription. RNase L destabilized the SRF transcript and formed a complex with SRF mRNA in cells providing a mechanism by which RNase L down-regulates SRF-induced genes. TTP and RNase L proteins interacted in cells suggesting that RNase L is directed to cleave TTP-bound RNAs as a mechanism of substrate specificity. Consistent with their concerted function in RNA turnover, the absence of either RNase L or TTP stabilized SRF mRNA, and a subset of established TTP targets was also regulated by RNase L. RNase L deficiency enhanced mitogen-induced proliferation demonstrating its functional role in limiting the mitogenic response. Our findings support a model of feedback regulation in which RNase L and TTP target SRF mRNA and SRF-induced transcripts. Accordingly, meta-analysis revealed an enrichment of RNase L and TTP targets among SRF-regulated genes suggesting that the RNase L/TTP axis represents a viable target to inhibit SRF-driven proliferation in neoplastic diseases.
Collapse
Affiliation(s)
- Sarah E Brennan-Laun
- From the Marlene and Stewart Greenebaum Cancer Center, Departments of Microbiology and Immunology and
| | - Xiao-Ling Li
- the Genetics Branch, NCI, National Institutes of Health, Bethesda, Maryland 20892
| | - Heather J Ezelle
- From the Marlene and Stewart Greenebaum Cancer Center, Departments of Microbiology and Immunology and the Research Services, Baltimore Veterans Affairs Medical Center, Baltimore, Maryland 21201, and
| | | | - Perry J Blackshear
- the Laboratory of Signal Transduction, NIEHS, National Institutes of Health, Research Triangle Park, North Carolina 27709
| | - Gerald M Wilson
- From the Marlene and Stewart Greenebaum Cancer Center, Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Bret A Hassel
- From the Marlene and Stewart Greenebaum Cancer Center, Departments of Microbiology and Immunology and the Research Services, Baltimore Veterans Affairs Medical Center, Baltimore, Maryland 21201, and
| |
Collapse
|
43
|
The neuronal activity-driven transcriptome. Mol Neurobiol 2014; 51:1071-88. [PMID: 24935719 DOI: 10.1007/s12035-014-8772-z] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Accepted: 06/01/2014] [Indexed: 10/25/2022]
Abstract
Activity-driven transcription is a key event associated with long-lasting forms of neuronal plasticity. Despite the efforts to investigate the regulatory mechanisms that control this complex process and the important advances in the knowledge of the function of many activity-induced genes in neurons, as well as the specific contribution of activity-regulated transcription factors, our understanding of how activity-driven transcription operates at the systems biology level is still very limited. This review focuses on the research of neuronal activity-driven transcription from an "omics" perspective. We will discuss the different high-throughput approaches undertaken to characterize the gene programs downstream of specific activity-regulated transcription factors, including CREB, SRF, MeCP2, Fos, Npas4, and others, and the interplay between epigenetic and transcriptional mechanisms underlying neuronal plasticity changes. Although basic questions remain unanswered and important challenges still lie ahead, the refinement of genome-wide techniques for investigating the neuronal transcriptome and epigenome promises great advances.
Collapse
|
44
|
The transcription factor serum response factor stimulates axon regeneration through cytoplasmic localization and cofilin interaction. J Neurosci 2014; 33:18836-48. [PMID: 24285890 DOI: 10.1523/jneurosci.3029-13.2013] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Axonal injury generates growth inert retraction bulbs with dynamic cytoskeletal properties that are severely compromised. Conversion of "frozen" retraction bulbs into actively progressing growth cones is a major aim in axon regeneration. Here we report that murine serum response factor (SRF), a gene regulator linked to the actin cytoskeleton, modulates growth cone actin dynamics during axon regeneration. In regeneration-competent facial motoneurons, Srf deletion inhibited axonal regeneration. In wild-type mice after nerve injury, SRF translocated from the nucleus to the cytoplasm, suggesting a cytoplasmic SRF function in axonal regeneration. Indeed, adenoviral overexpression of cytoplasmic SRF (SRF-ΔNLS-GFP) stimulated axonal sprouting and facial nerve regeneration in vivo. In primary central and peripheral neurons, SRF-ΔNLS-GFP stimulated neurite outgrowth, branch formation, and growth cone morphology. Furthermore, we uncovered a link between SRF and the actin-severing factor cofilin during axonal regeneration in vivo. Facial nerve axotomy increased the total cofilin abundance and also nuclear localization of phosphorylated cofilin in a subpopulation of lesioned motoneurons. This cytoplasmic-to-nucleus translocation of P-cofilin upon axotomy was reduced in motoneurons expressing SRF-ΔNLS-GFP. Finally, we demonstrate that cytoplasmic SRF and cofilin formed a reciprocal regulatory unit. Overexpression of cytoplasmic SRF reduced cofilin phosphorylation and vice versa: overexpression of cofilin inhibited SRF phosphorylation. Therefore, a regulatory loop consisting of SRF and cofilin might take part in reactivating actin dynamics in growth-inert retraction bulbs and facilitating axon regeneration.
Collapse
|
45
|
Schwann cells but not olfactory ensheathing cells inhibit CNS myelination via the secretion of connective tissue growth factor. J Neurosci 2014; 33:18686-97. [PMID: 24259589 DOI: 10.1523/jneurosci.3233-13.2013] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Cell transplantation is a promising strategy to promote CNS repair and has been studied for several decades with a focus on glial cells. Promising candidates include Schwann cells (SCs) and olfactory ensheathing cells (OECs). Both cell types are thought to be neural crest derived and share many properties in common, although OECs appear to be a better candidate for transplantation by evoking less astrogliosis. Using CNS mixed myelinating rat cultures plated on to a monolayer of astrocytes, we demonstrated that SCs, but not OECs, secrete a heat labile factor(s) that inhibits oligodendrocyte myelination. Comparative qRT-PCR and ELISA showed that SCs expressed higher levels of mRNA and protein for connective tissue growth factor (CTGF) than OECs. Anti-CTGF reversed the SCM-mediated effects on myelination. Both SCM and CTGF inhibited the differentiation of purified rat oligodendrocyte precursor cells (OPCs). Furthermore, pretreatment of astrocyte monolayers with SCM inhibited CNS myelination and led to transcriptional changes in the astrocyte, corresponding to upregulation of bone morphogenic protein 4 mRNA and CTGF mRNA (inhibitors of OPC differentiation) and the downregulation of insulin-like growth factor 2 mRNA (promoter of OPC differentiation). CTGF pretreatment of astrocytes increased their expression of CTGF, suggesting that this inhibitory factor can be positively regulated in astrocytes. These data provide evidence for the advantages of using OECs, and not mature SCs, for transplant-mediated repair and provide more evidence that they are a distinct and unique glial cell type.
Collapse
|
46
|
de Castro F, Bribián A, Ortega MC. Regulation of oligodendrocyte precursor migration during development, in adulthood and in pathology. Cell Mol Life Sci 2013; 70:4355-68. [PMID: 23689590 PMCID: PMC11113994 DOI: 10.1007/s00018-013-1365-6] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2012] [Revised: 05/03/2013] [Accepted: 05/07/2013] [Indexed: 12/25/2022]
Abstract
Oligodendrocytes are the myelin-forming cells in the central nervous system (CNS). These cells originate from oligodendrocyte precursor cells (OPCs) during development, and they migrate extensively from oligodendrogliogenic niches along the neural tube to colonise the entire CNS. Like many other such events, this migratory process is precisely regulated by a battery of positional and signalling cues that act via their corresponding receptors and that are expressed dynamically by OPCs. Here, we will review the cellular and molecular basis of this important event during embryonic and postnatal development, and we will discuss the relevance of the substantial number of OPCs existing in the adult CNS. Similarly, we will consider the behaviour of OPCs in normal and pathological conditions, especially in animal models of demyelination and of the demyelinating disease, multiple sclerosis. The spontaneous remyelination observed after damage in demyelinating pathologies has a limited effect. Understanding the cellular and molecular mechanisms underlying the biology of OPCs, particularly adult OPCs, should help in the design of neuroregenerative strategies to combat multiple sclerosis and other demyelinating diseases.
Collapse
Affiliation(s)
- Fernando de Castro
- Grupo de Neurobiología del Desarrollo-GNDe, Hospital Nacional de Parapléjicos-SESCAM, Finca "La Peraleda" s/n, 45071, Toledo, Spain,
| | | | | |
Collapse
|
47
|
Khodosevich K, Lazarini F, von Engelhardt J, Kaneko H, Lledo PM, Monyer H. Connective tissue growth factor regulates interneuron survival and information processing in the olfactory bulb. Neuron 2013; 79:1136-51. [PMID: 23993699 DOI: 10.1016/j.neuron.2013.07.011] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/11/2013] [Indexed: 12/27/2022]
Abstract
Neurogenesis underlies plastic changes in defined neuronal circuits in the postnatal and adult brain. Here we identify connective tissue growth factor (CTGF) as a critical factor in the mouse olfactory bulb (OB) in determining the efficiency of incorporation of postnatally born inhibitory neurons, thus gating the output of glomeruli, the first relay station of olfactory processing in the brain. In the OB, CTGF expression was restricted to prenatally born external tufted cells. CTGF enhanced the proapoptotic activity of glial-derived TGF-β2, decreasing the survival of periglomerular inhibitory neurons. Changes in CTGF expression levels in the OB led to modifications in local neuronal circuitry and olfactory behaviors. We show that the odorant-specific recruitment of distinct glomeruli resulted in enhanced local CTGF expression levels in the activated glomeruli. Collectively our data reveal a molecular mechanism controlling the survival of defined postnatally born neurons, thus adapting neuronal integration to the sensory experiences.
Collapse
Affiliation(s)
- Konstantin Khodosevich
- Department of Clinical Neurobiology, Heidelberg University Medical Center, 69120 Heidelberg, Germany; Department of Clinical Neurobiology/A230, German Center for Cancer Research (DKFZ), 69120 Heidelberg, Germany
| | | | | | | | | | | |
Collapse
|
48
|
Expression profiling of mouse subplate reveals a dynamic gene network and disease association with autism and schizophrenia. Proc Natl Acad Sci U S A 2013; 110:3555-60. [PMID: 23401504 DOI: 10.1073/pnas.1218510110] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
The subplate zone is a highly dynamic transient sector of the developing cerebral cortex that contains some of the earliest generated neurons and the first functional synapses of the cerebral cortex. Subplate cells have important functions in early establishment and maturation of thalamocortical connections, as well as in the development of inhibitory cortical circuits in sensory areas. So far no role has been identified for cells in the subplate in the mature brain and disease association of the subplate-specific genes has not been analyzed systematically. Here we present gene expression evidence for distinct roles of the mouse subplate across development as well as unique molecular markers to extend the repertoire of subplate labels. Performing systematic comparisons between different ages (embryonic days 15 and 18, postnatal day 8, and adult), we reveal the dynamic and constant features of the markers labeling subplate cells during embryonic and early postnatal development and in the adult. This can be visualized using the online database of subplate gene expression at https://molnar.dpag.ox.ac.uk/subplate/. We also identify embryonic similarities in gene expression between the ventricular zones, intermediate zone, and subplate, and distinct postnatal similarities between subplate, layer 5, and layers 2/3. The genes expressed in a subplate-specific manner at some point during development show a statistically significant enrichment for association with autism spectrum disorders and schizophrenia. Our report emphasizes the importance of the study of transient features of the developing brain to better understand neurodevelopmental disorders.
Collapse
|
49
|
Lin C, Hindes A, Burns CJ, Koppel AC, Kiss A, Yin Y, Ma L, Blumenberg M, Khnykin D, Jahnsen FL, Crosby SD, Ramanan N, Efimova T. Serum response factor controls transcriptional network regulating epidermal function and hair follicle morphogenesis. J Invest Dermatol 2012; 133:608-617. [PMID: 23151848 DOI: 10.1038/jid.2012.378] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Serum response factor (SRF) is a transcription factor that regulates the expression of growth-related immediate-early, cytoskeletal, and muscle-specific genes to control growth, differentiation, and cytoskeletal integrity in different cell types. To investigate the role for SRF in epidermal development and homeostasis, we conditionally knocked out SRF in epidermal keratinocytes. We report that SRF deletion disrupted epidermal barrier function leading to early postnatal lethality. Mice lacking SRF in epidermis displayed morphogenetic defects, including an eye-open-at-birth phenotype and lack of whiskers. SRF-null skin exhibited abnormal morphology, hyperplasia, aberrant expression of differentiation markers and transcriptional regulators, anomalous actin organization, enhanced inflammation, and retarded hair follicle (HF) development. Transcriptional profiling experiments uncovered profound molecular changes in SRF-null E17.5 epidermis and revealed that many previously identified SRF target CArG box-containing genes were markedly upregulated in SRF-null epidermis, indicating that SRF may function to repress transcription of a subset of its target genes in epidermis. Remarkably, when transplanted onto nude mice, engrafted SRF-null skin lacked hair but displayed normal epidermal architecture with proper expression of differentiation markers, suggesting that although keratinocyte SRF is essential for HF development, a cross-talk between SRF-null keratinocytes and the surrounding microenvironment is likely responsible for the barrier-deficient mutant epidermal phenotype.
Collapse
Affiliation(s)
- Congxing Lin
- Division of Dermatology, Department of Internal Medicine, Washington University School of Medicine, St Louis, Missouri, USA
| | - Anna Hindes
- Division of Dermatology, Department of Internal Medicine, Washington University School of Medicine, St Louis, Missouri, USA
| | - Carole J Burns
- Division of Dermatology, Department of Internal Medicine, Washington University School of Medicine, St Louis, Missouri, USA
| | - Aaron C Koppel
- Division of Dermatology, Department of Internal Medicine, Washington University School of Medicine, St Louis, Missouri, USA
| | - Alexi Kiss
- Division of Dermatology, Department of Internal Medicine, Washington University School of Medicine, St Louis, Missouri, USA
| | - Yan Yin
- Division of Dermatology, Department of Internal Medicine, Washington University School of Medicine, St Louis, Missouri, USA
| | - Liang Ma
- Division of Dermatology, Department of Internal Medicine, Washington University School of Medicine, St Louis, Missouri, USA
| | - Miroslav Blumenberg
- R. O. Perelman Department of Dermatology, NYU School of Medicine, New York, New York, USA
| | - Denis Khnykin
- Department of Pathology and Centre for Immune Regulation, University Hospital and University of Oslo, Oslo, Norway
| | - Frode L Jahnsen
- Department of Pathology and Centre for Immune Regulation, University Hospital and University of Oslo, Oslo, Norway
| | - Seth D Crosby
- Department of Genetics, Washington University School of Medicine, St Louis, Missouri, USA
| | - Narendrakumar Ramanan
- Department of Anatomy and Neurobiology, Washington University School of Medicine, St Louis, Missouri, USA
| | - Tatiana Efimova
- Division of Dermatology, Department of Internal Medicine, Washington University School of Medicine, St Louis, Missouri, USA.
| |
Collapse
|
50
|
Bouslama-Oueghlani L, Wehrlé R, Doulazmi M, Chen XR, Jaudon F, Lemaigre-Dubreuil Y, Rivals I, Sotelo C, Dusart I. Purkinje cell maturation participates in the control of oligodendrocyte differentiation: role of sonic hedgehog and vitronectin. PLoS One 2012; 7:e49015. [PMID: 23155445 PMCID: PMC3498367 DOI: 10.1371/journal.pone.0049015] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2011] [Accepted: 10/08/2012] [Indexed: 11/24/2022] Open
Abstract
Oligodendrocyte differentiation is temporally regulated during development by multiple factors. Here, we investigated whether the timing of oligodendrocyte differentiation might be controlled by neuronal differentiation in cerebellar organotypic cultures. In these cultures, the slices taken from newborn mice show very few oligodendrocytes during the first week of culture (immature slices) whereas their number increases importantly during the second week (mature slices). First, we showed that mature cerebellar slices or their conditioned media stimulated oligodendrocyte differentiation in immature slices thus demonstrating the existence of diffusible factors controlling oligodendrocyte differentiation. Using conditioned media from different models of slice culture in which the number of Purkinje cells varies drastically, we showed that the effects of these differentiating factors were proportional to the number of Purkinje cells. To identify these diffusible factors, we first performed a transcriptome analysis with an Affymetrix array for cerebellar cortex and then real-time quantitative PCR on mRNAs extracted from fluorescent flow cytometry sorted (FACS) Purkinje cells of L7-GFP transgenic mice at different ages. These analyses revealed that during postnatal maturation, Purkinje cells down-regulate Sonic Hedgehog and up-regulate vitronectin. Then, we showed that Sonic Hedgehog stimulates the proliferation of oligodendrocyte precursor cells and inhibits their differentiation. In contrast, vitronectin stimulates oligodendrocyte differentiation, whereas its inhibition with blocking antibodies abolishes the conditioned media effects. Altogether, these results suggest that Purkinje cells participate in controlling the timing of oligodendrocyte differentiation in the cerebellum through the developmentally regulated expression of diffusible molecules such as Sonic Hedgehog and vitronectin.
Collapse
Affiliation(s)
- Lamia Bouslama-Oueghlani
- Neurobiologie des Processus Adaptatif, Université Pierre et Marie Curie (UPMC Univ Paris 06), Paris, France
- Neurobiologie des Processus Adaptatif, CNRS (Centre National de Recherche Scientifique), Paris, France
| | - Rosine Wehrlé
- Neurobiologie des Processus Adaptatif, Université Pierre et Marie Curie (UPMC Univ Paris 06), Paris, France
- Neurobiologie des Processus Adaptatif, CNRS (Centre National de Recherche Scientifique), Paris, France
| | - Mohamed Doulazmi
- Neurobiologie des Processus Adaptatif, Université Pierre et Marie Curie (UPMC Univ Paris 06), Paris, France
- Neurobiologie des Processus Adaptatif, CNRS (Centre National de Recherche Scientifique), Paris, France
| | - Xiao Ru Chen
- Neurobiologie des Processus Adaptatif, Université Pierre et Marie Curie (UPMC Univ Paris 06), Paris, France
- Neurobiologie des Processus Adaptatif, CNRS (Centre National de Recherche Scientifique), Paris, France
| | - Fanny Jaudon
- Centre de Recherche de Biochimie Macromoléculaire, Université Montpellier 1 et 2, CNRS UMR 5237, Montpellier, France
| | - Yolande Lemaigre-Dubreuil
- Neurobiologie des Processus Adaptatif, Université Pierre et Marie Curie (UPMC Univ Paris 06), Paris, France
- Neurobiologie des Processus Adaptatif, CNRS (Centre National de Recherche Scientifique), Paris, France
| | - Isabelle Rivals
- Équipe de statistique Appliquée, ESPCI ParisTech (Ecole Supérieure de Physique et Chimie Industrielles de la Ville de Paris), Paris, France
| | - Constantino Sotelo
- Neurobiologie des Processus Adaptatif, Université Pierre et Marie Curie (UPMC Univ Paris 06), Paris, France
- Neurobiologie des Processus Adaptatif, CNRS (Centre National de Recherche Scientifique), Paris, France
- Instituto de Neurociencias, Universidad Miguel Hernández–CSIC, San Juan de Alicante, Spain
| | - Isabelle Dusart
- Neurobiologie des Processus Adaptatif, Université Pierre et Marie Curie (UPMC Univ Paris 06), Paris, France
- Neurobiologie des Processus Adaptatif, CNRS (Centre National de Recherche Scientifique), Paris, France
- * E-mail:
| |
Collapse
|