1
|
Attwell CL, Maldonado-Lasunción I, Eggers R, Bijleveld BA, Ellenbroek WM, Siersema N, Razenberg L, Lamme D, Fagoe ND, van Kesteren RE, Smit AB, Verhaagen J, Mason MRJ. The transcription factor combination MEF2 and KLF7 promotes axonal sprouting in the injured spinal cord with functional improvement and regeneration-associated gene expression. Mol Neurodegener 2025; 20:18. [PMID: 39923113 PMCID: PMC11807332 DOI: 10.1186/s13024-025-00805-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 01/23/2025] [Indexed: 02/10/2025] Open
Abstract
BACKGROUND Axon regeneration after injury to the central nervous system (CNS) is limited by an inhibitory environment but also because injured neurons fail to initiate expression of regeneration associated genes (RAGs). The potential of strong RAG expression to promote regeneration in the CNS is exemplified by the conditioning lesion model, whereby peripheral nerve injury promotes regeneration of centrally projecting branches of the injured neurons. RAG expression could potentially be induced by delivery of the right set of transcription factors (TFs). We here aim to identify TF combinations that activate this program. METHODS We first analysed binding site motifs in promoters of the RAG program to identify nine candidate growth-promoting TFs. These were systematically screened in vitro to identify combinations that had potent neurite-growth promoting activity. Next, adeno-associated viral vectors were used to express these TF combinations in vivo in L4/L5 dorsal root ganglia to test whether they would promote regeneration in a spinal cord injury model (dorsal column lesion) in female rats. To determine whether they could activate the RAG program we carried out gene expression profiling on laser-dissected dorsal root ganglion neurons specifically expressing these TF combinations, and of DRG neurons that had been axotomized. RESULTS Promoter analysis identified ATF3, Jun, CEBPD, KLF7, MEF2, SMAD1, SOX11, STAT3 and SRF as candidate RAG-activating TFs. In vitro screening identified two TF combinations, KLF7/MEF2 and ATF3/KLF7/MEF2, that had potent neurite-growth promoting activity, the latter being the more powerful. In vivo, KLF7/MEF2, but not ATF3/KLF7/MEF2 or KLF7 or MEF2 alone, promoted axonal sprouting into the dorsal column lesion site and led to improved functional recovery. Gene expression profiling revealed that unexpectedly, the MEF2-VP16 construct used had little transcriptional activity in vivo, suggesting additional steps may be required to achieve full MEF2 activity. All combinations except MEF2 alone induced RAG expression mirroring that induced by axotomy to significant extents, while ATF3/KLF7/MEF2, KLF7 and ATF3, but not KLF7/MEF2 also induced apoptosis-related genes which may hinder regeneration. CONCLUSIONS The TF combination KLF7/MEF2 partially mimics the conditioning lesion effect, inducing axonal sprouting into a dorsal column lesion and driving significant RAG expression, and also promotes functional improvement.
Collapse
Affiliation(s)
- Callan L Attwell
- Laboratory for Regeneration of Sensorimotor Systems, Netherlands Institute for Neuroscience, An Institute of the Royal Academy of Arts and Sciences, Amsterdam, the Netherlands
| | - Inés Maldonado-Lasunción
- Laboratory for Regeneration of Sensorimotor Systems, Netherlands Institute for Neuroscience, An Institute of the Royal Academy of Arts and Sciences, Amsterdam, the Netherlands
- Division of Neuroscience, Department of Brain Sciences, Imperial College London, London, W12 0NN, UK
| | - Ruben Eggers
- Laboratory for Regeneration of Sensorimotor Systems, Netherlands Institute for Neuroscience, An Institute of the Royal Academy of Arts and Sciences, Amsterdam, the Netherlands
| | - Bastiaan A Bijleveld
- Laboratory for Regeneration of Sensorimotor Systems, Netherlands Institute for Neuroscience, An Institute of the Royal Academy of Arts and Sciences, Amsterdam, the Netherlands
| | - Ward M Ellenbroek
- Laboratory for Regeneration of Sensorimotor Systems, Netherlands Institute for Neuroscience, An Institute of the Royal Academy of Arts and Sciences, Amsterdam, the Netherlands
| | - Natascha Siersema
- Laboratory for Regeneration of Sensorimotor Systems, Netherlands Institute for Neuroscience, An Institute of the Royal Academy of Arts and Sciences, Amsterdam, the Netherlands
| | - Lotte Razenberg
- Laboratory for Regeneration of Sensorimotor Systems, Netherlands Institute for Neuroscience, An Institute of the Royal Academy of Arts and Sciences, Amsterdam, the Netherlands
| | - Dédé Lamme
- Laboratory for Regeneration of Sensorimotor Systems, Netherlands Institute for Neuroscience, An Institute of the Royal Academy of Arts and Sciences, Amsterdam, the Netherlands
| | - Nitish D Fagoe
- Laboratory for Regeneration of Sensorimotor Systems, Netherlands Institute for Neuroscience, An Institute of the Royal Academy of Arts and Sciences, Amsterdam, the Netherlands
| | - Ronald E van Kesteren
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognition Research, Neuroscience Campus Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - August B Smit
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognition Research, Neuroscience Campus Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Joost Verhaagen
- Laboratory for Regeneration of Sensorimotor Systems, Netherlands Institute for Neuroscience, An Institute of the Royal Academy of Arts and Sciences, Amsterdam, the Netherlands
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognition Research, Neuroscience Campus Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Matthew R J Mason
- Laboratory for Regeneration of Sensorimotor Systems, Netherlands Institute for Neuroscience, An Institute of the Royal Academy of Arts and Sciences, Amsterdam, the Netherlands.
| |
Collapse
|
2
|
Ma P, Duan S, Ma W, Deng Q, Yu Y, Gao P, Yuan Y, Liu C. Single-cell chromatin accessibility landscape profiling reveals the diversity of epigenetic regulation in the rat nervous system. Sci Data 2025; 12:140. [PMID: 39856121 PMCID: PMC11761061 DOI: 10.1038/s41597-025-04432-y] [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: 08/01/2024] [Accepted: 01/06/2025] [Indexed: 01/27/2025] Open
Abstract
The mammalian nervous system controls complex functions through highly specialized and interacting structures. Single-cell sequencing can provide information on cell-type-specific chromatin structure and regulatory elements, revealing differences in chromatin organization between different cell types and their potential roles of these differences in brain function. Here, we generated a chromatin accessibility dataset through single-cell ATAC-seq of 174,593 high-quality nuclei from 16 adult rat brain regions. We identified cell subtypes of both neuronal and non-neuronal cells with highly specific distributions and characterized gene regulatory elements associated with cell type-specific regions. To further investigate the gene regulatory network involved in spinal cord regeneration, we integrated our scATAC-seq data with published single-nucleus RNA-seq data from the spinal cord, and we identified more detailed regeneration related elements by drawing GRNs centered on the transcription factor Jun in the OPC. We also performed similar integration analyses in the midbrain. Our findings provide a solid foundation for the comprehensive dissection of the molecular architecture of the mammalian nervous system.
Collapse
Affiliation(s)
- Peiyao Ma
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006, China
- BGI Research, Hangzhou, 310030, China
| | - Shanshan Duan
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
- BGI Research, Hangzhou, 310030, China
| | - Wen Ma
- BGI Research, Hangzhou, 310030, China
| | - Qiuting Deng
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
- BGI Research, Hangzhou, 310030, China
| | - Yeya Yu
- BGI Research, Hangzhou, 310030, China
| | - Peng Gao
- Shanxi Medical University-BGI Collaborative Center for Future Medicine, Shanxi Medical University, Taiyuan, 030001, China
- BGI, Shenzhen, 518083, China
| | - Yue Yuan
- BGI Research, Hangzhou, 310030, China.
- BGI Research, Shenzhen, 518083, China.
| | - Chuanyu Liu
- BGI Research, Shenzhen, 518083, China.
- Shanxi Medical University-BGI Collaborative Center for Future Medicine, Shanxi Medical University, Taiyuan, 030001, China.
- Shenzhen Proof-of-Concept Center of Digital Cytopathology, BGI Research, Shenzhen, 518083, China.
- Key Laboratory of Immune Mechanism and Intervention on Serious Disease in Hebei Province, Department of Immunology, Hebei Medical University, Shijiazhuang, 050017, China.
| |
Collapse
|
3
|
Tang H, Pan J, Xu Y, Liu L, Yang X, Huang S, Peng T, Huang Y, Zhao Y, Fu C, Zhou H, Chen Z, Wang W, He L, Xu K. Constraint therapy promotes motor cortex remodeling and functional improvement by regulating c-Jun/miR-182-5p/Nogo - A signals in hemiplegic cerebral palsy mice. Ann Anat 2023; 250:152136. [PMID: 37506776 DOI: 10.1016/j.aanat.2023.152136] [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: 12/14/2022] [Revised: 05/18/2023] [Accepted: 07/04/2023] [Indexed: 07/30/2023]
Abstract
BACKGROUND Our previous study has confirmed that constraint-induced movement therapy (CIMT) could promote neural remodeling in hemiplegic cerebral palsy (HCP) mice through Nogo-A/NgR/RhoA/ROCK signaling, however, the upstream mechanism was still unclear. Therefore, the present study aimed to further explore the mechanism of CIMT regulating the expression of Nogo-A in HCP mice. METHOD HCP mice were well established through ligating the left common carotid artery of 7-day-old pups and being placed in a hypoxic box which was filled with a mixture of 8% oxygen and 92% nitrogen. CIMT intervention was conducted by taping to fix the entire arm of the contralateral side (left) to force the mice to use the affected limb (right). Bioinformatics prediction and luciferase experiment were performed to confirm that miR-182-5p was targeted with Nogo-A. The beam test and grip test were applied to examine the behavioral performance under the intervention of c-Jun and CIMT. Also, immunofluorescence, Golgi staining, and transmission electron microscopy were conducted to show that the lenti-expression of c-Jun could increases the expression of myelin, and downregulates the expression of Nogo-A under the CIMT on HCP mice. RESULT (1) The beam walking test and grip test experiment results showed that compared with the control group, the HCP + nCIMT group's forelimb grasping ability and balance coordination ability were decreased (P < 0.05). (2) The results of Golgi staining, and transmission electron microscopy showed that the thickness of myelin sheath and the density of dendritic spines in the HCP + nCIMT group were lower than those in the control group (P < 0.05). Compared with the HCP + nCIMT group, the cerebral cortex myelin sheath thickness, dendrite spine density and nerve filament expression were increased in HCP + CIMT group (P < 0.05). (3) Immunofluorescence staining showed that the expression of Nogo-A in the cerebral cortex of the HCP + nCIMT group was higher than that of the HCP + CIMT group (P < 0.05). Compared with the HCP + CIMT group, the expression of Nogo-A in the HCP + LC + CIMT group was decreased and, in the HCP, + SC + CIMT group was significantly increased (P < 0.05). Compared with the HCP + nCIMT group, the expression of c-Jun in the control, HCP + CIMT, HCP + LC + nCIMT and HCP + LC + CIMT groups was significantly increased, and in the HCP + SC + CIMT was decreased (P < 0.05). (4) Real-time quantitative polymerase chain reaction (RT-qPCR) results showed that the expression level of miR-182-5p in the HCP + LC + CIMT group was more increased than that in the HCP + nCIMT group (P < 0.05). The expression level of miR-182-5p in the HCP + LC + CIMT group was higher than that in the HCP + LC + nCIMT group and the HCP + SC + CIMT group (P < 0.05). CONCLUSION These data identified that CIMT might stimulate the remodeling of neurons and myelin in the motor cortex by partially inhibiting the c-Jun/miR-182-5p/Nogo-A pathway, thereby facilitating the grasping performance and balance function of HCP mice.
Collapse
Affiliation(s)
- Hongmei Tang
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jing Pan
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yunxian Xu
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Liru Liu
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xubo Yang
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Shiya Huang
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Tingting Peng
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yuan Huang
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yiting Zhao
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Chaoqiong Fu
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Hongyu Zhou
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zhaofang Chen
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Wenda Wang
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Lu He
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Kaishou Xu
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
4
|
Zhu Y, Luan C, Gong L, Gu Y, Wang X, Sun H, Chen Z, Zhou Q, Liu C, Shan Q, Gu X, Zhou S. SnRNA-seq reveals the heterogeneity of spinal ventral horn and mechanism of motor neuron axon regeneration. iScience 2023; 26:107264. [PMID: 37502257 PMCID: PMC10368823 DOI: 10.1016/j.isci.2023.107264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 05/02/2023] [Accepted: 06/27/2023] [Indexed: 07/29/2023] Open
Abstract
Spinal motor neurons, the distinctive neurons of the central nervous system, extend into the peripheral nervous system and have outstanding ability of axon regeneration after injury. Here, we explored the heterogeneity of spinal ventral horn cells after rat sciatic nerve crush via single-nuclei RNA sequencing. Interestingly, regeneration mainly occurred in a Sncg+ and Anxa2+ motor neuron subtype (MN2) surrounded by a newly emerged microglia subtype (Mg6) after injury. Subsequently, microglia depletion slowed down the regeneration of sciatic nerve. OPCs were also involved into the regeneration process. Knockdown of Cacna2d2 in vitro and systemic blocking of Cacna2d2 in vivo improved the axon growth ability, hinting us the importance of Ca2+. Ultimately, we proposed three possible phases of motor neuron axon regeneration: preparation stage, early regeneration stage, and regeneration stage. Taken together, our study provided a resource for deciphering the underlying mechanism of motor neuron axon regeneration in a single cell dimension.
Collapse
Affiliation(s)
- Ye Zhu
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300000, China
| | - Chengcheng Luan
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300000, China
| | - Leilei Gong
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu 226001, China
| | - Yun Gu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu 226001, China
| | - Xinghui Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu 226001, China
| | - Hualin Sun
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu 226001, China
| | - Zhifeng Chen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu 226001, China
| | - Qiang Zhou
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu 226001, China
| | - Chang Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu 226001, China
| | - Qi Shan
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300000, China
| | - Xiaosong Gu
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300000, China
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu 226001, China
| | - Songlin Zhou
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu 226001, China
| |
Collapse
|
5
|
Lear BP, Moore DL. Moving CNS axon growth and regeneration research into human model systems. Front Neurosci 2023; 17:1198041. [PMID: 37425013 PMCID: PMC10324669 DOI: 10.3389/fnins.2023.1198041] [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: 03/31/2023] [Accepted: 05/25/2023] [Indexed: 07/11/2023] Open
Abstract
Axon regeneration is limited in the adult mammalian central nervous system (CNS) due to both intrinsic and extrinsic factors. Rodent studies have shown that developmental age can drive differences in intrinsic axon growth ability, such that embryonic rodent CNS neurons extend long axons while postnatal and adult CNS neurons do not. In recent decades, scientists have identified several intrinsic developmental regulators in rodents that modulate growth. However, whether this developmentally programmed decline in CNS axon growth is conserved in humans is not yet known. Until recently, there have been limited human neuronal model systems, and even fewer age-specific human models. Human in vitro models range from pluripotent stem cell-derived neurons to directly reprogrammed (transdifferentiated) neurons derived from human somatic cells. In this review, we discuss the advantages and disadvantages of each system, and how studying axon growth in human neurons can provide species-specific knowledge in the field of CNS axon regeneration with the goal of bridging basic science studies to clinical trials. Additionally, with the increased availability and quality of 'omics datasets of human cortical tissue across development and lifespan, scientists can mine these datasets for developmentally regulated pathways and genes. As there has been little research performed in human neurons to study modulators of axon growth, here we provide a summary of approaches to begin to shift the field of CNS axon growth and regeneration into human model systems to uncover novel drivers of axon growth.
Collapse
Affiliation(s)
| | - Darcie L. Moore
- Department of Neuroscience, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
6
|
Maxson Jones K, Morgan JR. Lampreys and spinal cord regeneration: "a very special claim on the interest of zoologists," 1830s-present. Front Cell Dev Biol 2023; 11:1113961. [PMID: 37228651 PMCID: PMC10203415 DOI: 10.3389/fcell.2023.1113961] [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: 12/01/2022] [Accepted: 04/24/2023] [Indexed: 05/27/2023] Open
Abstract
Employing history of science methods, including analyses of the scientific literature, archival documents, and interviews with scientists, this paper presents a history of lampreys in neurobiology from the 1830s to the present. We emphasize the lamprey's roles in helping to elucidate spinal cord regeneration mechanisms. Two attributes have long perpetuated studies of lampreys in neurobiology. First, they possess large neurons, including multiple classes of stereotypically located, 'identified' giant neurons in the brain, which project their large axons into the spinal cord. These giant neurons and their axonal fibers have facilitated electrophysiological recordings and imaging across biological scales, ranging from molecular to circuit-level analyses of nervous system structures and functions and including their roles in behavioral output. Second, lampreys have long been considered amongst the most basal extant vertebrates on the planet, so they have facilitated comparative studies pointing to conserved and derived characteristics of vertebrate nervous systems. These features attracted neurologists and zoologists to studies of lampreys between the 1830s and 1930s. But, the same two attributes also facilitated the rise of the lamprey in neural regeneration research after 1959, when biologists first wrote about the spontaneous, robust regeneration of some identified CNS axons in larvae after spinal cord injuries, coupled with recovery of normal swimming. Not only did large neurons promote fresh insights in the field, enabling studies incorporating multiple scales with existing and new technologies. But investigators also were able to attach a broad scope of relevance to their studies, interpreting them as suggesting conserved features of successful, and sometimes even unsuccessful, CNS regeneration. Lamprey research demonstrated that functional recovery takes place without the reformation of the original neuronal connections, for instance, by way of imperfect axonal regrowth and compensatory plasticity. Moreover, research performed in the lamprey model revealed that factors intrinsic to neurons are integral in promoting or hindering regeneration. As this work has helped illuminate why basal vertebrates accomplish CNS regeneration so well, whereas mammals do it so poorly, this history presents a case study in how biological and medical value have been, and could continue to be, gleaned from a non-traditional model organism for which molecular tools have been developed only relatively recently.
Collapse
Affiliation(s)
- Kathryn Maxson Jones
- Center for Medical Ethics and Health Policy, Baylor College of Medicine, Houston, TX, United States
- Department of History, Purdue University, West Lafayette, IN, United States
- Marine Biological Laboratory, The Eugene Bell Center for Regenerative Biology and Tissue Engineering, Woods Hole, MA, United States
| | - Jennifer R. Morgan
- Marine Biological Laboratory, The Eugene Bell Center for Regenerative Biology and Tissue Engineering, Woods Hole, MA, United States
| |
Collapse
|
7
|
Schwab K, Coronel L, Riege K, Sacramento EK, Rahnis N, Häckes D, Cirri E, Groth M, Hoffmann S, Fischer M. Multi-omics analysis identifies RFX7 targets involved in tumor suppression and neuronal processes. Cell Death Discov 2023; 9:80. [PMID: 36864036 PMCID: PMC9981735 DOI: 10.1038/s41420-023-01378-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 02/19/2023] [Accepted: 02/22/2023] [Indexed: 03/04/2023] Open
Abstract
Recurrently mutated in lymphoid neoplasms, the transcription factor RFX7 is emerging as a tumor suppressor. Previous reports suggested that RFX7 may also have a role in neurological and metabolic disorders. We recently reported that RFX7 responds to p53 signaling and cellular stress. Furthermore, we found RFX7 target genes to be dysregulated in numerous cancer types also beyond the hematological system. However, our understanding of RFX7's target gene network and its role in health and disease remains limited. Here, we generated RFX7 knock-out cells and employed a multi-omics approach integrating transcriptome, cistrome, and proteome data to obtain a more comprehensive picture of RFX7 targets. We identify novel target genes linked to RFX7's tumor suppressor function and underscoring its potential role in neurological disorders. Importantly, our data reveal RFX7 as a mechanistic link that enables the activation of these genes in response to p53 signaling.
Collapse
Affiliation(s)
- Katjana Schwab
- grid.418245.e0000 0000 9999 5706Computational Biology Group, Leibniz Institute on Aging—Fritz Lipmann Institute (FLI), Beutenbergstraße 11, 07745 Jena, Germany
| | - Luis Coronel
- grid.418245.e0000 0000 9999 5706Computational Biology Group, Leibniz Institute on Aging—Fritz Lipmann Institute (FLI), Beutenbergstraße 11, 07745 Jena, Germany
| | - Konstantin Riege
- grid.418245.e0000 0000 9999 5706Computational Biology Group, Leibniz Institute on Aging—Fritz Lipmann Institute (FLI), Beutenbergstraße 11, 07745 Jena, Germany
| | - Erika K. Sacramento
- grid.418245.e0000 0000 9999 5706Core Facility for Proteomics, Leibniz Institute on Aging—Fritz Lipmann Institute (FLI), Beutenbergstraße 11, 07745 Jena, Germany
| | - Norman Rahnis
- grid.418245.e0000 0000 9999 5706Core Facility for Proteomics, Leibniz Institute on Aging—Fritz Lipmann Institute (FLI), Beutenbergstraße 11, 07745 Jena, Germany
| | - David Häckes
- grid.418245.e0000 0000 9999 5706Computational Biology Group, Leibniz Institute on Aging—Fritz Lipmann Institute (FLI), Beutenbergstraße 11, 07745 Jena, Germany
| | - Emilio Cirri
- grid.418245.e0000 0000 9999 5706Core Facility for Proteomics, Leibniz Institute on Aging—Fritz Lipmann Institute (FLI), Beutenbergstraße 11, 07745 Jena, Germany
| | - Marco Groth
- grid.418245.e0000 0000 9999 5706Core Facility for Next-Generation Sequencing, Leibniz Institute on Aging—Fritz Lipmann Institute (FLI), Beutenbergstraße 11, 07745 Jena, Germany
| | - Steve Hoffmann
- grid.418245.e0000 0000 9999 5706Computational Biology Group, Leibniz Institute on Aging—Fritz Lipmann Institute (FLI), Beutenbergstraße 11, 07745 Jena, Germany
| | - Martin Fischer
- Computational Biology Group, Leibniz Institute on Aging-Fritz Lipmann Institute (FLI), Beutenbergstraße 11, 07745, Jena, Germany.
| |
Collapse
|
8
|
Construction and analysis of mRNA, lncRNA, and transcription factor regulatory networks after retinal ganglion cell injury. Exp Eye Res 2021; 215:108915. [PMID: 34971620 DOI: 10.1016/j.exer.2021.108915] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Revised: 12/11/2021] [Accepted: 12/23/2021] [Indexed: 12/11/2022]
Abstract
Retinal ganglion cell (RGC) injury is a critical pathological feature of several optic neurodegenerative diseases. The regulatory mechanisms underlying RGC injury remain poorly understood. Recent evidence has highlighted the important roles of long noncoding RNAs (lncRNAs) in degenerative neuropathy but few studies have focused on lncRNAs associated with RGC injury. In this study, we analyzed dysregulated lncRNAs associated with RGC injury, their potential regulatory functions, and the molecular mechanisms underlying the regulation of lncRNAs and transcription factors (TFs). We analyzed lncRNA and mRNA profiles in the GSE142881 dataset associated with RGC injury and identified 1049 differentially expressed genes (DEGs), with 18 differentially expressed (DE) TFs among 883 DE mRNAs and 312 DE lncRNAs. The predicted DE lncRNAs and DE mRNAs were used to construct a lncRNA-mRNA co-expression network. Functional enrichment analysis was performed to explore the functions of the lncRNAs and mRNAs. The co-expression network between DE lncRNAs and DE mRNAs was highly enriched in inflammatory and immune-related pathways, indicating that they play role in the process of RGC injury. Among the DE mRNAs, we screened 18 DE TFs, including activating transcription factor 3 (ATF3), associated with RGC injury. Co-expression analysis predicted that 13 lncRNAs were potential binding targets of ATF3. The screening of the potential targets of these 13 lncRNAs showed that they were also significantly enriched in functional pathways associated with inflammation and apoptosis. After analysis, we constructed the mRNA-ATF3-lncRNA regulatory network after RGCs injury. In summary, we identified the gene module associated with immune and inflammatory responses after optic nerve injury and constructed a regulatory network of lncRNA-TF-mRNA. The results indicate that lncRNAs, by binding to TFs, can regulate downstream genes and function during RGC injury. The results provide a foundation for further studies of the mechanism of RGC injury and provide insight into the clinical diagnosis and investigation direction of neurodegenerative diseases such as traumatic optic neuropathy and glaucoma.
Collapse
|
9
|
Campion TJ, Sheikh IS, Smit RD, Iffland PH, Chen J, Junker IP, Krynska B, Crino PB, Smith GM. Viral expression of constitutively active AKT3 induces CST axonal sprouting and regeneration, but also promotes seizures. Exp Neurol 2021; 349:113961. [PMID: 34953897 DOI: 10.1016/j.expneurol.2021.113961] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 12/17/2021] [Accepted: 12/18/2021] [Indexed: 12/01/2022]
Abstract
Increasing the intrinsic growth potential of neurons after injury has repeatedly been shown to promote some level of axonal regeneration in rodent models. One of the most studied pathways involves the activation of the PI3K/AKT/mTOR pathways, primarily by reducing the levels of PTEN, a negative regulator of PI3K. Likewise, activation of signal transducer and activator of transcription 3 (STAT3) has previously been shown to boost axonal regeneration and sprouting within the injured nervous system. Here, we examined the regeneration of the corticospinal tract (CST) after cortical expression of constitutively active (ca) Akt3 and STAT3, both separately and in combination. Overexpression of caAkt3 induced regeneration of CST axons past the injury site independent of caSTAT3 overexpression. STAT3 demonstrated improved axon sprouting compared to controls and contributed to a synergistic improvement in effects when combined with Akt3 but failed to promote axonal regeneration as an individual therapy. Despite showing impressive axonal regeneration, animals expressing Akt3 failed to show any functional improvement and deteriorated with time. During this period, we observed progressive Akt3 dose-dependent increase in behavioral seizures. Histology revealed increased phosphorylation of ribosomal S6 protein within the unilateral cortex, increased neuronal size, microglia activation and hemispheric enlargement (hemimegalencephaly).
Collapse
Affiliation(s)
- Thomas J Campion
- Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, 3500 North Broad Street, Philadelphia, PA 19140, United States of America; Shriners Hospitals Pediatric Research Center, Lewis Katz School of Medicine, Temple University, 3500 North Broad Street, Philadelphia, PA 19140, United States of America
| | - Imran S Sheikh
- Shriners Hospitals Pediatric Research Center, Lewis Katz School of Medicine, Temple University, 3500 North Broad Street, Philadelphia, PA 19140, United States of America
| | - Rupert D Smit
- Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, 3500 North Broad Street, Philadelphia, PA 19140, United States of America; Shriners Hospitals Pediatric Research Center, Lewis Katz School of Medicine, Temple University, 3500 North Broad Street, Philadelphia, PA 19140, United States of America
| | - Philip H Iffland
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Jie Chen
- Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, 3500 North Broad Street, Philadelphia, PA 19140, United States of America; Shriners Hospitals Pediatric Research Center, Lewis Katz School of Medicine, Temple University, 3500 North Broad Street, Philadelphia, PA 19140, United States of America
| | - Ian P Junker
- Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, 3500 North Broad Street, Philadelphia, PA 19140, United States of America
| | - Barbara Krynska
- Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, 3500 North Broad Street, Philadelphia, PA 19140, United States of America; Shriners Hospitals Pediatric Research Center, Lewis Katz School of Medicine, Temple University, 3500 North Broad Street, Philadelphia, PA 19140, United States of America
| | - Peter B Crino
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - George M Smith
- Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, 3500 North Broad Street, Philadelphia, PA 19140, United States of America; Shriners Hospitals Pediatric Research Center, Lewis Katz School of Medicine, Temple University, 3500 North Broad Street, Philadelphia, PA 19140, United States of America.
| |
Collapse
|
10
|
Lowell JA, O’Neill N, Danzi MC, Al-Ali H, Bixby JL, Lemmon VP. Phenotypic Screening Following Transcriptomic Deconvolution to Identify Transcription Factors Mediating Axon Growth Induced by a Kinase Inhibitor. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2021; 26:1337-1354. [PMID: 34218704 PMCID: PMC10509783 DOI: 10.1177/24725552211026270] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
After injury to the central nervous system (CNS), both neuron-intrinsic limitations on regenerative responses and inhibitory factors in the injured CNS environment restrict regenerative axon growth. Instances of successful axon regrowth offer opportunities to identify features that differentiate these situations from that of the normal adult CNS. One such opportunity is provided by the kinase inhibitor RO48, which dramatically enhances neurite outgrowth of neurons in vitro and substantially increased contralateral sprouting of corticospinal tract neurons when infused intraventricularly following unilateral pyramidotomy. The authors present here a transcriptomic deconvolution of RO48-associated axon growth, with the goal of identifying transcriptional regulators associated with axon growth in the CNS. Through the use of RNA sequencing (RNA-seq) and transcription factor binding site enrichment analysis, the authors identified a list of transcription factors putatively driving differential gene expression during RO48-induced neurite outgrowth of rat hippocampal neurons in vitro. The 82 transcription factor motifs identified in this way included some with known association to axon growth regulation, such as Jun, Klf4, Myc, Atf4, Stat3, and Nfatc2, and many with no known association to axon growth. A phenotypic loss-of-function screen was carried out to evaluate these transcription factors for their roles in neurite outgrowth; this screen identified several potential outgrowth regulators. Subsequent validation suggests that the Forkhead box (Fox) family transcription factor Foxp2 restricts neurite outgrowth, while FoxO subfamily members Foxo1 and Foxo3a promote neurite outgrowth. The authors' combined transcriptomic-phenotypic screening strategy therefore allowed identification of novel transcriptional regulators of neurite outgrowth downstream of a multitarget kinase inhibitor.
Collapse
Affiliation(s)
- Jeffrey A. Lowell
- Miami Project to Cure Paralysis and University of Miami Miller School of Medicine, Miami, FL, USA
| | - Nicholas O’Neill
- Miami Project to Cure Paralysis and University of Miami Miller School of Medicine, Miami, FL, USA
| | - Matt C. Danzi
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Hassan Al-Ali
- Miami Project to Cure Paralysis and University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Medicine and Peggy & Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, FL, USA
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - John L. Bixby
- Miami Project to Cure Paralysis and University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL, USA
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
- Dr. John T. Macdonald Foundation Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Vance P. Lemmon
- Miami Project to Cure Paralysis and University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL, USA
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
11
|
Venkatesh I, Mehra V, Wang Z, Simpson MT, Eastwood E, Chakraborty A, Beine Z, Gross D, Cabahug M, Olson G, Blackmore MG. Co-occupancy identifies transcription factor co-operation for axon growth. Nat Commun 2021; 12:2555. [PMID: 33953205 PMCID: PMC8099911 DOI: 10.1038/s41467-021-22828-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 03/29/2021] [Indexed: 12/13/2022] Open
Abstract
Transcription factors (TFs) act as powerful levers to regulate neural physiology and can be targeted to improve cellular responses to injury or disease. Because TFs often depend on cooperative activity, a major challenge is to identify and deploy optimal sets. Here we developed a bioinformatics pipeline, centered on TF co-occupancy of regulatory DNA, and used it to predict factors that potentiate the effects of pro-regenerative Klf6 in vitro. High content screens of neurite outgrowth identified cooperative activity by 12 candidates, and systematic testing in a mouse model of corticospinal tract (CST) damage substantiated three novel instances of pairwise cooperation. Combined Klf6 and Nr5a2 drove the strongest growth, and transcriptional profiling of CST neurons identified Klf6/Nr5a2-responsive gene networks involved in macromolecule biosynthesis and DNA repair. These data identify TF combinations that promote enhanced CST growth, clarify the transcriptional correlates, and provide a bioinformatics approach to detect TF cooperation.
Collapse
Affiliation(s)
- Ishwariya Venkatesh
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI, USA.
| | - Vatsal Mehra
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI, USA
| | - Zimei Wang
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI, USA
| | - Matthew T Simpson
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI, USA
| | - Erik Eastwood
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI, USA
| | | | - Zac Beine
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI, USA
| | - Derek Gross
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI, USA
| | - Michael Cabahug
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI, USA
| | - Greta Olson
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI, USA
| | - Murray G Blackmore
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI, USA.
| |
Collapse
|
12
|
Lee N, Lee SH, Lee J, Lee MY, Lim J, Kim S, Kim S. Hepatocyte growth factor is necessary for efficient outgrowth of injured peripheral axons in in vitro culture system and in vivo nerve crush mouse model. Biochem Biophys Rep 2021; 26:100973. [PMID: 33718632 PMCID: PMC7933716 DOI: 10.1016/j.bbrep.2021.100973] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 02/18/2021] [Accepted: 02/22/2021] [Indexed: 11/12/2022] Open
Abstract
Hepatocyte growth factor (HGF) is a neurotrophic factor and its role in peripheral nerves has been relatively unknown. In this study, biological functions of HGF and its receptor c-met have been investigated in the context of regeneration of damaged peripheral nerves. Axotomy of the peripheral branch of sensory neurons from embryonic dorsal root ganglia (DRG) resulted in the increased protein levels of HGF and phosphorylated c-met. When the neuronal cultures were treated with a pharmacological inhibitor of c-met, PHA665752, the length of axotomy-induced outgrowth of neurite was significantly reduced. On the other hand, the addition of recombinant HGF proteins to the neuronal culture facilitated axon outgrowth. In the nerve crush mouse model, the protein level of HGF was increased around the injury site by almost 5.5-fold at 24 h post injury compared to control mice and was maintained at elevated levels for another 6 days. The amount of phosphorylated c-met receptor in sciatic nerve was also observed to be higher than control mice. When PHA665752 was locally applied to the injury site of sciatic nerve, axon outgrowth and injury mediated induction of cJun protein were effectively inhibited, indicating the functional involvement of HGF/c-met pathway in the nerve regeneration process. When extra HGF was exogenously provided by intramuscular injection of plasmid DNA expressing HGF, axon outgrowth from damaged sciatic nerve and cJun expression level were enhanced. Taken together, these results suggested that HGF/c-met pathway plays important roles in axon outgrowth by directly interacting with sensory neurons and thus HGF might be a useful tool for developing therapeutics for peripheral neuropathy. In in vitro primary eDRGs, axotomy-induced HGF/c-met pathway enhanced the neurite outgrowth process. Nerve injury induced the expression of HGF, consequently leading to the activation of c-met in peripheral axons. HGF/c-met pathway played an important role in the regeneration process of injured peripheral nerves. Additional supply of HGF, in the form of plasmid DNA, enhanced the regeneration of damaged peripheral nerves.
Collapse
Affiliation(s)
- Nayeon Lee
- School of Biological Sciences, Seoul National University, Seoul, 08826, South Korea.,Division of Gene Therapy, Helixmith Co Ltd, Seoul, 07794, South Korea
| | - Sang Hwan Lee
- School of Biological Sciences, Seoul National University, Seoul, 08826, South Korea
| | - Junghun Lee
- Division of Gene Therapy, Helixmith Co Ltd, Seoul, 07794, South Korea
| | - Mi-Young Lee
- Division of Gene Therapy, Helixmith Co Ltd, Seoul, 07794, South Korea
| | - Jaegook Lim
- Division of Gene Therapy, Helixmith Co Ltd, Seoul, 07794, South Korea
| | - Subin Kim
- Division of Gene Therapy, Helixmith Co Ltd, Seoul, 07794, South Korea
| | - Sunyoung Kim
- Division of Gene Therapy, Helixmith Co Ltd, Seoul, 07794, South Korea
| |
Collapse
|
13
|
Role of Myc Proto-Oncogene as a Transcriptional Hub to Regulate the Expression of Regeneration-Associated Genes following Preconditioning Peripheral Nerve Injury. J Neurosci 2021; 41:446-460. [PMID: 33262248 DOI: 10.1523/jneurosci.1745-20.2020] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 11/16/2020] [Accepted: 11/18/2020] [Indexed: 12/20/2022] Open
Abstract
Preconditioning peripheral nerve injury enhances the intrinsic growth capacity of DRGs sensory axons by inducing transcriptional upregulation of the regeneration-associated genes (RAGs). However, it is still unclear how preconditioning injury leads to the orchestrated induction of many RAGs. The present study identified Myc proto-oncogene as a transcriptional hub gene to regulate the expression of a distinct subset of RAGs in DRGs following the preconditioning injury. We demonstrated that c-MYC bound to the promoters of certain RAGs, such as Jun, Atf3, and Sprr1a, and that Myc upregulation following SNI preceded that of the RAGs bound by c-MYC. Marked DNA methylation of the Myc exon 3 sequences was implicated in the early transcriptional activation and accompanied by open histone marks. Myc deletion led to a decrease in the injury-induced expression of a distinct subset of RAGs, which were highly overlapped with the list of RAGs that were upregulated by Myc overexpression. Following dorsal hemisection spinal cord injury in female rats, Myc overexpression in DRGs significantly prevented the retraction of the sensory axons in a manner dependent on its downstream RAG, June Our results suggest that Myc plays a critical role in axon regeneration via its transcriptional activity to regulate the expression of a spectrum of downstream RAGs and subsequent effector molecules. Identification of more upstream hub transcription factors and the epigenetic mechanisms specific for individual hub transcription factors would advance our understanding of how the preconditioning injury induces orchestrated upregulation of RAGs.
Collapse
|
14
|
Abstract
Permanent disabilities following CNS injuries result from the failure of injured axons to regenerate and rebuild functional connections with their original targets. By contrast, injury to peripheral nerves is followed by robust regeneration, which can lead to recovery of sensory and motor functions. This regenerative response requires the induction of widespread transcriptional and epigenetic changes in injured neurons. Considerable progress has been made in recent years in understanding how peripheral axon injury elicits these widespread changes through the coordinated actions of transcription factors, epigenetic modifiers and, to a lesser extent, microRNAs. Although many questions remain about the interplay between these mechanisms, these new findings provide important insights into the pivotal role of coordinated gene expression and chromatin remodelling in the neuronal response to injury.
Collapse
Affiliation(s)
- Marcus Mahar
- Department of Neuroscience, Hope Center for Neurological Disorders and Center of Regenerative Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Valeria Cavalli
- Department of Neuroscience, Hope Center for Neurological Disorders and Center of Regenerative Medicine, Washington University School of Medicine, St Louis, MO, USA.
| |
Collapse
|
15
|
Bayramov AV, Ermakova GV, Kucheryavyy AV, Zaraisky AG. Lampreys, “Living Fossils,” in Research on Early Development and Regeneration in Vertebrates. Russ J Dev Biol 2019. [DOI: 10.1134/s1062360418080015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
16
|
Venkatesh I, Mehra V, Wang Z, Califf B, Blackmore MG. Developmental Chromatin Restriction of Pro-Growth Gene Networks Acts as an Epigenetic Barrier to Axon Regeneration in Cortical Neurons. Dev Neurobiol 2018; 78:960-977. [PMID: 29786967 PMCID: PMC6204296 DOI: 10.1002/dneu.22605] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 05/01/2018] [Accepted: 05/04/2018] [Indexed: 12/21/2022]
Abstract
Axon regeneration in the central nervous system is prevented in part by a developmental decline in the intrinsic regenerative ability of maturing neurons. This loss of axon growth ability likely reflects widespread changes in gene expression, but the mechanisms that drive this shift remain unclear. Chromatin accessibility has emerged as a key regulatory mechanism in other cellular contexts, raising the possibility that chromatin structure may contribute to the age-dependent loss of regenerative potential. Here we establish an integrated bioinformatic pipeline that combines analysis of developmentally dynamic gene networks with transcription factor regulation and genome-wide maps of chromatin accessibility. When applied to the developing cortex, this pipeline detected overall closure of chromatin in sub-networks of genes associated with axon growth. We next analyzed mature CNS neurons that were supplied with various pro-regenerative transcription factors. Unlike prior results with SOX11 and KLF7, here we found that neither JUN nor an activated form of STAT3 promoted substantial corticospinal tract regeneration. Correspondingly, chromatin accessibility in JUN or STAT3 target genes was substantially lower than in predicted targets of SOX11 and KLF7. Finally, we used the pipeline to predict pioneer factors that could potentially relieve chromatin constraints at growth-associated loci. Overall this integrated analysis substantiates the hypothesis that dynamic chromatin accessibility contributes to the developmental decline in axon growth ability and influences the efficacy of pro-regenerative interventions in the adult, while also pointing toward selected pioneer factors as high-priority candidates for future combinatorial experiments. © 2018 Wiley Periodicals, Inc. Develop Neurobiol 00: 000-000, 2018.
Collapse
Affiliation(s)
| | - Vatsal Mehra
- Department of Biomedical Sciences, Marquette University, 53201
| | - Zimei Wang
- Department of Biomedical Sciences, Marquette University, 53201
| | | | | |
Collapse
|
17
|
Danzi MC, Mehta ST, Dulla K, Zunino G, Cooper DJ, Bixby JL, Lemmon VP. The effect of Jun dimerization on neurite outgrowth and motif binding. Mol Cell Neurosci 2018; 92:114-127. [PMID: 30077771 PMCID: PMC6547139 DOI: 10.1016/j.mcn.2018.08.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 07/31/2018] [Accepted: 08/01/2018] [Indexed: 12/20/2022] Open
Abstract
Axon regeneration is a necessary step toward functional recovery after spinal cord injury. The AP-1 transcription factor c-Jun has long been known to play an important role in directing the transcriptional response of Dorsal Root Ganglion (DRG) neurons to peripheral axotomy that results in successful axon regeneration. Here we performed ChIPseq for Jun in mouse DRG neurons after a sciatic nerve crush or sham surgery in order to measure the changes in Jun's DNA binding in response to peripheral axotomy. We found that the majority of Jun's injury-responsive changes in DNA binding occur at putative enhancer elements, rather than proximal to transcription start sites. We also used a series of single polypeptide chain tandem transcription factors to test the effects of different Jun-containing dimers on neurite outgrowth in DRG, cortical and hippocampal neurons. These experiments demonstrated that dimers composed of Jun and Atf3 promoted neurite outgrowth in rat CNS neurons as well as mouse DRG neurons. Our work provides new insight into the mechanisms underlying Jun's role in axon regeneration.
Collapse
Affiliation(s)
- Matt C Danzi
- Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, USA; Center for Computational Science, University of Miami, Miami, FL, USA; Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Saloni T Mehta
- Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, USA; Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Kireeti Dulla
- Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Giulia Zunino
- Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, USA; Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Daniel J Cooper
- Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, USA; Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - John L Bixby
- Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, USA; Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA; Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL, USA.
| | - Vance P Lemmon
- Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, USA; Center for Computational Science, University of Miami, Miami, FL, USA; Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
18
|
KLF6 and STAT3 co-occupy regulatory DNA and functionally synergize to promote axon growth in CNS neurons. Sci Rep 2018; 8:12565. [PMID: 30135567 PMCID: PMC6105645 DOI: 10.1038/s41598-018-31101-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 08/10/2018] [Indexed: 11/26/2022] Open
Abstract
The failure of axon regeneration in the CNS limits recovery from damage and disease. Members of the KLF family of transcription factors can exert both positive and negative effects on axon regeneration, but the underlying mechanisms are unclear. Here we show that forced expression of KLF6 promotes axon regeneration by corticospinal tract neurons in the injured spinal cord. RNA sequencing identified 454 genes whose expression changed upon forced KLF6 expression in vitro, including sub-networks that were highly enriched for functions relevant to axon extension including cytoskeleton remodeling, lipid synthesis, and bioenergetics. In addition, promoter analysis predicted a functional interaction between KLF6 and a second transcription factor, STAT3, and genome-wide footprinting using ATAC-Seq data confirmed frequent co-occupancy. Co-expression of the two factors yielded a synergistic elevation of neurite growth in vitro. These data clarify the transcriptional control of axon growth and point the way toward novel interventions to promote CNS regeneration.
Collapse
|
19
|
Rheaume BA, Jereen A, Bolisetty M, Sajid MS, Yang Y, Renna K, Sun L, Robson P, Trakhtenberg EF. Single cell transcriptome profiling of retinal ganglion cells identifies cellular subtypes. Nat Commun 2018; 9:2759. [PMID: 30018341 PMCID: PMC6050223 DOI: 10.1038/s41467-018-05134-3] [Citation(s) in RCA: 268] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 06/12/2018] [Indexed: 12/13/2022] Open
Abstract
Retinal ganglion cells (RGCs) convey the major output of information collected from the eye to the brain. Thirty subtypes of RGCs have been identified to date. Here, we analyze 6225 RGCs (average of 5000 genes per cell) from right and left eyes by single-cell RNA-seq and classify them into 40 subtypes using clustering algorithms. We identify additional subtypes and markers, as well as transcription factors predicted to cooperate in specifying RGC subtypes. Zic1, a marker of the right eye-enriched subtype, is validated by immunostaining in situ. Runx1 and Fst, the markers of other subtypes, are validated in purified RGCs by fluorescent in situ hybridization (FISH) and immunostaining. We show the extent of gene expression variability needed for subtype segregation, and we show a hierarchy in diversification from a cell-type population to subtypes. Finally, we present a website for comparing the gene expression of RGC subtypes.
Collapse
Affiliation(s)
- Bruce A Rheaume
- Department of Neuroscience, University of Connecticut School of Medicine, 263 Farmington Ave, Farmington, CT, 06030, USA
| | - Amyeo Jereen
- Department of Neuroscience, University of Connecticut School of Medicine, 263 Farmington Ave, Farmington, CT, 06030, USA
| | - Mohan Bolisetty
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, 06032, USA
| | - Muhammad S Sajid
- Department of Neuroscience, University of Connecticut School of Medicine, 263 Farmington Ave, Farmington, CT, 06030, USA
| | - Yue Yang
- Department of Neuroscience, University of Connecticut School of Medicine, 263 Farmington Ave, Farmington, CT, 06030, USA
| | - Kathleen Renna
- Department of Neuroscience, University of Connecticut School of Medicine, 263 Farmington Ave, Farmington, CT, 06030, USA
| | - Lili Sun
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, 06032, USA
| | - Paul Robson
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, 06032, USA
- Institute for Systems Genomics and Department of Genetics & Genome Sciences, University of Connecticut School of Medicine, Farmington, CT, 06032, USA
| | - Ephraim F Trakhtenberg
- Department of Neuroscience, University of Connecticut School of Medicine, 263 Farmington Ave, Farmington, CT, 06030, USA.
| |
Collapse
|
20
|
Ghosh S, Hui SP. Axonal regeneration in zebrafish spinal cord. REGENERATION (OXFORD, ENGLAND) 2018; 5:43-60. [PMID: 29721326 PMCID: PMC5911453 DOI: 10.1002/reg2.99] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 03/09/2018] [Accepted: 03/13/2018] [Indexed: 12/12/2022]
Abstract
In the present review we discuss two interrelated events-axonal damage and repair-known to occur after spinal cord injury (SCI) in the zebrafish. Adult zebrafish are capable of regenerating axonal tracts and can restore full functionality after SCI. Unlike fish, axon regeneration in the adult mammalian central nervous system is extremely limited. As a consequence of an injury there is very little repair of disengaged axons and therefore functional deficit persists after SCI in adult mammals. In contrast, peripheral nervous system axons readily regenerate following injury and hence allow functional recovery both in mammals and fish. A better mechanistic understanding of these three scenarios could provide a more comprehensive insight into the success or failure of axonal regeneration after SCI. This review summarizes the present understanding of the cellular and molecular basis of axonal regeneration, in both the peripheral nervous system and the central nervous system, and large scale gene expression analysis is used to focus on different events during regeneration. The discovery and identification of genes involved in zebrafish spinal cord regeneration and subsequent functional experimentation will provide more insight into the endogenous mechanism of myelination and remyelination. Furthermore, precise knowledge of the mechanism underlying the extraordinary axonal regeneration process in zebrafish will also allow us to unravel the potential therapeutic strategies to be implemented for enhancing regrowth and remyelination of axons in mammals.
Collapse
Affiliation(s)
- Sukla Ghosh
- Department of BiophysicsMolecular Biology and BioinformaticsUniversity of Calcutta92 A. P. C. RoadKolkata 700009India
| | - Subhra Prakash Hui
- Department of BiophysicsMolecular Biology and BioinformaticsUniversity of Calcutta92 A. P. C. RoadKolkata 700009India
- Victor Chang Cardiac Research InstituteLowy Packer Building, 405 Liverpool StDarlinghurstNSW 2010Australia.
| |
Collapse
|
21
|
Shen Q, Yasmeen R, Marbourg J, Xu L, Yu L, Fadda P, Flechtner A, Lee LJ, Popovich PG, Ziouzenkova O. Induction of innervation by encapsulated adipocytes with engineered vitamin A metabolism. Transl Res 2018; 192:1-14. [PMID: 29144959 PMCID: PMC5811336 DOI: 10.1016/j.trsl.2017.10.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 09/26/2017] [Accepted: 10/13/2017] [Indexed: 12/13/2022]
Abstract
Innervation is a fundamental basis for function and survival of tissues. In the peripheral tissues, degenerative diseases create a neurotoxic metabolic milieu that either causes neurodegeneration or fails to sustain regenerative growth and reinnervation of injured/diseased tissues. Encapsulation of cells producing neurotrophic factors can augment axon growth and neuron survival; however, sustained innervation in vivo requires a combination of factors promoting axon growth and guidance pathway that are released in a tissue-specific context. Using novel encapsulation techniques and genetic tools, we manipulated retinoic acid-generating enzyme aldehyde dehydrogenase 1a1 (Aldh1a1) in adipocytes that are capable of promoting growth and innervation of white adipose tissue by sympathetic neurons. Aldh1a1-/- adipocytes secrete molecules that regulate axon guidance and markedly stimulate neurite outgrowth in vitro and in vivo. Based on studies with natural and synthetic RAR agonists and antagonists, gene microarray and nanostring arrays, we concluded that ephrin A5/ephrin A4 is a downstream pathway regulated by Aldh1a1. Encapsulation of Aldh1a1-/- adipocytes into alginate poly-L-lysine microcapsules induced functional innervation of adipose tissue in obese wild-type mice. We propose that encapsulated Aldh1a1-/- adipocytes could provide a therapeutic solution for the reinnervation of damaged tissues.
Collapse
Affiliation(s)
- Qiwen Shen
- Department of Human Sciences, The Ohio State University, Columbus, Ohio
| | - Rumana Yasmeen
- Department of Human Sciences, The Ohio State University, Columbus, Ohio
| | - Jessica Marbourg
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, Ohio; Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, Ohio
| | - Lu Xu
- Department of Human Sciences, The Ohio State University, Columbus, Ohio; Department of Minimally Invasive Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Lianbo Yu
- Department of Statistics, The Ohio State University College of Medicine, Columbus, Ohio
| | - Paolo Fadda
- Nucleic Acid Shared Resource, Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Alan Flechtner
- Histology and Immunohistochemistry Laboratory, The Ohio State University, Columbus, Ohio
| | - L James Lee
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio
| | - Phillip G Popovich
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, Ohio; Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, Ohio
| | | |
Collapse
|
22
|
Herman PE, Papatheodorou A, Bryant SA, Waterbury CKM, Herdy JR, Arcese AA, Buxbaum JD, Smith JJ, Morgan JR, Bloom O. Highly conserved molecular pathways, including Wnt signaling, promote functional recovery from spinal cord injury in lampreys. Sci Rep 2018; 8:742. [PMID: 29335507 PMCID: PMC5768751 DOI: 10.1038/s41598-017-18757-1] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 12/18/2017] [Indexed: 12/23/2022] Open
Abstract
In mammals, spinal cord injury (SCI) leads to dramatic losses in neurons and synaptic connections, and consequently function. Unlike mammals, lampreys are vertebrates that undergo spontaneous regeneration and achieve functional recovery after SCI. Therefore our goal was to determine the complete transcriptional responses that occur after SCI in lampreys and to identify deeply conserved pathways that promote regeneration. We performed RNA-Seq on lamprey spinal cord and brain throughout the course of functional recovery. We describe complex transcriptional responses in the injured spinal cord, and somewhat surprisingly, also in the brain. Transcriptional responses to SCI in lampreys included transcription factor networks that promote peripheral nerve regeneration in mammals such as Atf3 and Jun. Furthermore, a number of highly conserved axon guidance, extracellular matrix, and proliferation genes were also differentially expressed after SCI in lampreys. Strikingly, ~3% of differentially expressed transcripts belonged to the Wnt pathways. These included members of the Wnt and Frizzled gene families, and genes involved in downstream signaling. Pharmacological inhibition of Wnt signaling inhibited functional recovery, confirming a critical role for this pathway. These data indicate that molecular signals present in mammals are also involved in regeneration in lampreys, supporting translational relevance of the model.
Collapse
Affiliation(s)
- Paige E Herman
- The Feinstein Institute for Medical Research, Center for Autoimmune and Musculoskeletal Disease, Manhasset, NY, 11030, USA
| | - Angelos Papatheodorou
- The Feinstein Institute for Medical Research, Center for Autoimmune and Musculoskeletal Disease, Manhasset, NY, 11030, USA
| | - Stephanie A Bryant
- University of Kentucky, Department of Biology, Lexington, KY, 40506, USA
| | | | - Joseph R Herdy
- University of Kentucky, Department of Biology, Lexington, KY, 40506, USA
| | - Anthony A Arcese
- The Feinstein Institute for Medical Research, Center for Autoimmune and Musculoskeletal Disease, Manhasset, NY, 11030, USA
| | - Joseph D Buxbaum
- Icahn School of Medicine at Mount Sinai, Department of Psychiatry, New York, NY, 10029, USA
| | - Jeramiah J Smith
- University of Kentucky, Department of Biology, Lexington, KY, 40506, USA
| | - Jennifer R Morgan
- Marine Biological Laboratory, The Eugene Bell Center for Regenerative Biology and Tissue Engineering, Woods Hole, MA, 02543, USA.
| | - Ona Bloom
- The Feinstein Institute for Medical Research, Center for Autoimmune and Musculoskeletal Disease, Manhasset, NY, 11030, USA.
| |
Collapse
|
23
|
High Content Screening of Mammalian Primary Cortical Neurons. Methods Mol Biol 2017. [PMID: 29082499 DOI: 10.1007/978-1-4939-7357-6_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
High Content Screening (HCS) can be used to analyze the morphology of neuronal primary cultures on a large scale. When used in the field of neuronal regeneration this approach allows the screening of hundreds or thousands of perturbagens, such as miRNAs, cDNAs, or compounds, for their ability to induce neuronal growth. One of the most important steps while designing these kinds of experiments is the choice of the correct neuronal model. Testing the correct neuronal type is critical to obtain results that are biologically significant and that can later be translated to a clinical setting. For example, if the goal is identifying possible therapies for Spinal Cord Injury (SCI), a challenging target is the neuronal projection from the motor cortex to the spinal cord, the corticospinal tract. Here, we describe the experimental protocols that can be used to produce primary cortical culture from young rat cortices, electroporate the neurons to study the effect of altered gene expression on neurite growth, and immunostain to measure neurite growth parameters.
Collapse
|
24
|
Kaplan A, Bueno M, Hua L, Fournier AE. Maximizing functional axon repair in the injured central nervous system: Lessons from neuronal development. Dev Dyn 2017. [PMID: 28643358 DOI: 10.1002/dvdy.24536] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
The failure of damaged axons to regrow underlies disability in central nervous system injury and disease. Therapies that stimulate axon repair will be critical to restore function. Extensive axon regeneration can be induced by manipulation of oncogenes and tumor suppressors; however, it has been difficult to translate this into functional recovery in models of spinal cord injury. The current challenge is to maximize the functional integration of regenerating axons to recover motor and sensory behaviors. Insights into axonal growth and wiring during nervous system development are helping guide new approaches to boost regeneration and functional connectivity after injury in the mature nervous system. Here we discuss our current understanding of axonal behavior after injury and prospects for the development of drugs to optimize axon regeneration and functional recovery after CNS injury. Developmental Dynamics 247:18-23, 2018. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Andrew Kaplan
- Department of Neurology and Neurosurgery, Montréal Neurological Institute, McGill University, Montréal, Québec, Canada
| | - Mardja Bueno
- Department of Neurology and Neurosurgery, Montréal Neurological Institute, McGill University, Montréal, Québec, Canada
| | - Luyang Hua
- Department of Neurology and Neurosurgery, Montréal Neurological Institute, McGill University, Montréal, Québec, Canada
| | - Alyson E Fournier
- Department of Neurology and Neurosurgery, Montréal Neurological Institute, McGill University, Montréal, Québec, Canada
| |
Collapse
|
25
|
Motti D, Lerch JK, Danzi MC, Gans JH, Kuo F, Slepak TI, Bixby JL, Lemmon VP. Identification of miRNAs involved in DRG neurite outgrowth and their putative targets. FEBS Lett 2017; 591:2091-2105. [PMID: 28626869 PMCID: PMC5864114 DOI: 10.1002/1873-3468.12718] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 06/07/2017] [Accepted: 06/11/2017] [Indexed: 12/14/2022]
Abstract
Peripheral neurons regenerate their axons after injury. Transcriptional regulation by microRNAs (miRNAs) is one possible mechanism controlling regeneration. We profiled miRNA expression in mouse dorsal root ganglion neurons after a sciatic nerve crush, and identified 49 differentially expressed miRNAs. We evaluated the functional role of each miRNA using a phenotypic analysis approach. To predict the targets of the miRNAs we employed RNA-Sequencing and examined transcription at the isoform level. We identify thousands of differentially expressed isoforms and bioinformatically associate the miRNAs that modulate neurite growth with their putative target isoforms to outline a network of regulatory events underlying peripheral nerve regeneration. MiR-298, let-7a, and let-7f enhance neurite growth and target the majority of isoforms in the differentially expressed network.
Collapse
Affiliation(s)
- Dario Motti
- The Miami Project To Cure Paralysis, The University of Miami Miller School of Medicine, Miami, FL
| | - Jessica K. Lerch
- The Department of Neuroscience, The Ohio State University, Columbus, OH
| | - Matt C. Danzi
- The Miami Project To Cure Paralysis, The University of Miami Miller School of Medicine, Miami, FL
| | - Jared H. Gans
- The Miami Project To Cure Paralysis, The University of Miami Miller School of Medicine, Miami, FL
| | - Frank Kuo
- The Miami Project To Cure Paralysis, The University of Miami Miller School of Medicine, Miami, FL
| | - Tatiana I. Slepak
- The Miami Project To Cure Paralysis, The University of Miami Miller School of Medicine, Miami, FL
| | - John L. Bixby
- The Miami Project To Cure Paralysis, The University of Miami Miller School of Medicine, Miami, FL
- The Department of Molecular and Cellular Pharmacology, The University of Miami Miller School of Medicine, Miami, FL
- The Department of Neurological Surgery, The University of Miami Miller School of Medicine, Miami, FL
- The Center for Computational Science, The University of Miami, Miami, FL
| | - Vance P. Lemmon
- The Miami Project To Cure Paralysis, The University of Miami Miller School of Medicine, Miami, FL
- The Department of Neurological Surgery, The University of Miami Miller School of Medicine, Miami, FL
- The Center for Computational Science, The University of Miami, Miami, FL
| |
Collapse
|
26
|
Mehta ST, Bixby JL, Lemmon VP. Genetically modifying transcription factors to promote CNS axon regeneration. Neural Regen Res 2017; 12:737-738. [PMID: 28616023 PMCID: PMC5461604 DOI: 10.4103/1673-5374.206637] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Affiliation(s)
- Saloni T Mehta
- Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, USA
| | - John L Bixby
- Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, USA.,Center for Computational Science, University of Miami Miller School of Medicine, Miami, FL, USA.,Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA.,Molecular & Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Vance P Lemmon
- Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, USA.,Center for Computational Science, University of Miami Miller School of Medicine, Miami, FL, USA.,Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
27
|
Rinaldi F, Motti D, Ferraiuolo L, Kaspar BK. High content analysis in amyotrophic lateral sclerosis. Mol Cell Neurosci 2017; 80:180-191. [PMID: 27965018 PMCID: PMC5393940 DOI: 10.1016/j.mcn.2016.12.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 12/05/2016] [Accepted: 12/09/2016] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating disease characterized by the progressive loss of motor neurons. Neurons, astrocytes, oligodendrocytes and microglial cells all undergo pathological modifications in the onset and progression of ALS. A number of genes involved in the etiopathology of the disease have been identified, but a complete understanding of the molecular mechanisms of ALS has yet to be determined. Currently, people affected by ALS have a life expectancy of only two to five years from diagnosis. The search for a treatment has been slow and mostly unsuccessful, leaving patients in desperate need of better therapies. Until recently, most pre-clinical studies utilized the available ALS animal models. In the past years, the development of new protocols for isolation of patient cells and differentiation into relevant cell types has provided new tools to model ALS, potentially more relevant to the disease itself as they directly come from patients. The use of stem cells is showing promise to facilitate ALS research by expanding our understanding of the disease and help to identify potential new therapeutic targets and therapies to help patients. Advancements in high content analysis (HCA) have the power to contribute to move ALS research forward by combining automated image acquisition along with digital image analysis. With modern HCA machines it is possible, in a period of just a few hours, to observe changes in morphology and survival of cells, under the stimulation of hundreds, if not thousands of drugs and compounds. In this article, we will summarize the major molecular and cellular hallmarks of ALS, describe the advancements provided by the in vitro models developed in the last few years, and review the studies that have applied HCA to the ALS field to date.
Collapse
Affiliation(s)
- Federica Rinaldi
- Center for Gene Therapy, Nationwide Children's Hospital, Columbus, OH, USA
| | - Dario Motti
- Center for Gene Therapy, Nationwide Children's Hospital, Columbus, OH, USA
| | - Laura Ferraiuolo
- Center for Gene Therapy, Nationwide Children's Hospital, Columbus, OH, USA; Department of Neuroscience, Sheffield Institute of Translational Neuroscience, University of Sheffield, UK
| | - Brian K Kaspar
- Center for Gene Therapy, Nationwide Children's Hospital, Columbus, OH, USA; Department of Neuroscience, The Ohio State University, Columbus, OH, USA; Department of Pediatrics, College of Medicine and Public Health, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
28
|
Cooper DJ, Zunino G, Bixby JL, Lemmon VP. Phenotypic screening with primary neurons to identify drug targets for regeneration and degeneration. Mol Cell Neurosci 2017; 80:161-169. [PMID: 27444126 PMCID: PMC5243932 DOI: 10.1016/j.mcn.2016.07.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 07/04/2016] [Accepted: 07/16/2016] [Indexed: 12/13/2022] Open
Abstract
High-throughput, target-based screening techniques have been utilized extensively for drug discovery in the past several decades. However, the need for more predictive in vitro models of in vivo disease states has generated a shift in strategy towards phenotype-based screens. Phenotype based screens are particularly valuable in studying complex conditions such as CNS injury and degenerative disease, as many factors can contribute to a specific cellular response. In this review, we will discuss different screening frameworks and their relative utility in examining mechanisms of neurodegeneration and axon regrowth, particularly in cell-based in vitro disease models.
Collapse
Affiliation(s)
- Daniel J. Cooper
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami, 1400 NW 12th Ave, Miami, FL 33136, USA
| | - Giulia Zunino
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami, 1400 NW 12th Ave, Miami, FL 33136, USA
| | - John L. Bixby
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami, 1400 NW 12th Ave, Miami, FL 33136, USA
- Center for Computational Science, University of Miami, 1400 NW 12th Ave, Miami, FL 33136, USA
- Department of Molecular and Cellular Pharmacology, Miller School of Medicine, University of Miami, 1400 NW 12th Ave, Miami, FL 33136, USA
| | - Vance P. Lemmon
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami, 1400 NW 12th Ave, Miami, FL 33136, USA
- Center for Computational Science, University of Miami, 1400 NW 12th Ave, Miami, FL 33136, USA
| |
Collapse
|
29
|
Callif BL, Maunze B, Krueger NL, Simpson MT, Blackmore MG. The application of CRISPR technology to high content screening in primary neurons. Mol Cell Neurosci 2017; 80:170-179. [PMID: 28110021 DOI: 10.1016/j.mcn.2017.01.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 01/09/2017] [Accepted: 01/17/2017] [Indexed: 10/20/2022] Open
Abstract
Axon growth is coordinated by multiple interacting proteins that remain incompletely characterized. High content screening (HCS), in which manipulation of candidate genes is combined with rapid image analysis of phenotypic effects, has emerged as a powerful technique to identify key regulators of axon outgrowth. Here we explore the utility of a genome editing approach referred to as CRISPR (Clustered Regularly Interspersed Palindromic Repeats) for knockout screening in primary neurons. In the CRISPR approach a DNA-cleaving Cas enzyme is guided to genomic target sequences by user-created guide RNA (sgRNA), where it initiates a double-stranded break that ultimately results in frameshift mutation and loss of protein production. Using electroporation of plasmid DNA that co-expresses Cas9 enzyme and sgRNA, we first verified the ability of CRISPR targeting to achieve protein-level knockdown in cultured postnatal cortical neurons. Targeted proteins included NeuN (RbFox3) and PTEN, a well-studied regulator of axon growth. Effective knockdown lagged at least four days behind transfection, but targeted proteins were eventually undetectable by immunohistochemistry in >80% of transfected cells. Consistent with this, anti-PTEN sgRNA produced no changes in neurite outgrowth when assessed three days post-transfection. When week-long cultures were replated, however, PTEN knockdown consistently increased neurite lengths. These CRISPR-mediated PTEN effects were achieved using multi-well transfection and automated phenotypic analysis, indicating the suitability of PTEN as a positive control for future CRISPR-based screening efforts. Combined, these data establish an example of CRISPR-mediated protein knockdown in primary cortical neurons and its compatibility with HCS workflows.
Collapse
Affiliation(s)
- Ben L Callif
- Department of Biomedical Sciences, Marquette University, 53201, USA
| | - Brian Maunze
- Department of Biomedical Sciences, Marquette University, 53201, USA
| | - Nick L Krueger
- Department of Biomedical Sciences, Marquette University, 53201, USA
| | | | | |
Collapse
|
30
|
Venkatesh I, Simpson MT, Coley DM, Blackmore MG. Epigenetic profiling reveals a developmental decrease in promoter accessibility during cortical maturation in vivo. NEUROEPIGENETICS 2016; 8:19-26. [PMID: 27990351 PMCID: PMC5159751 DOI: 10.1016/j.nepig.2016.10.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Axon regeneration in adult central nervous system (CNS) is limited in part by a developmental decline in the ability of injured neurons to re-express needed regeneration associated genes (RAGs). Adult CNS neurons may lack appropriate pro-regenerative transcription factors, or may display chromatin structure that restricts transcriptional access to RAGs. Here we performed epigenetic profiling around the promoter regions of key RAGs, and found progressive restriction across a time course of cortical maturation. These data identify a potential intrinsic constraint to axon growth in adult CNS neurons. Neurite outgrowth from cultured postnatal cortical neurons, however, proved insensitive to treatments that improve axon growth in other cell types, including combinatorial overexpression of AP1 factors, overexpression of histone acetyltransferases, and pharmacological inhibitors of histone deacetylases. This insensitivity could be due to intermediate chromatin closure at the time of culture, and highlights important differences in cell culture models used to test potential pro-regenerative interventions.
Collapse
Affiliation(s)
| | | | - Denise M. Coley
- Department of Biomedical Sciences, Marquette University, 53201
| | | |
Collapse
|
31
|
Zhang Q, Zhang ZJ, Wang XH, Ma J, Song YH, Liang M, Lin SX, Zhao J, Zhang AZ, Li F, Hua Q. The prescriptions from Shenghui soup enhanced neurite growth and GAP-43 expression level in PC12 cells. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2016; 16:369. [PMID: 27646829 PMCID: PMC5029060 DOI: 10.1186/s12906-016-1339-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 09/05/2016] [Indexed: 11/28/2022]
Abstract
Background Shenghui soup is a traditional Chinese herbal medicine used in clinic for the treatment of forgetfulness. In order to understanding the prescription principle, the effects of “tonifying qi and strengthening spleen” group (TQSS) including Poria cocos (Schw.) Wolf. and Panax ginseng C.A.Mey and “eliminating phlegm and strengthening intelligence” group (EPSI) composed of Polygala tenuifolia Willd., Acorus calamus L. and Sinapis alba L from the herb complex on neurite growth in PC12 cells, two disassembled prescriptions derived from Shenghui soup and their molecular mechanisms were investigated. Methods Firstly, CCK-8 kit was used to detect the impact of the two prescriptions on PC12 cell viability; and Flow cytometry was performed to measure the cell apoptosis when PC12 cells were treated with these drugs. Secondly, the effect of the two prescriptions on the differentiation of PC12 cells was observed. Finally, the mRNA and protein expression levels of GAP-43 were analyzed by RT-PCR and western blot, respectively. Results “Tonifying qi and strengthening spleen” prescription decreased cell viability in a dose-dependent manner, but had no significant effect on cell apoptosis. Meanwhile, it could improve neurite growth and elevate the mRNA and protein expression level of GAP-43. “Eliminating phlegm and strengthening intelligence” prescription also exerted the similar effects on cell viability and apoptosis. Furthermore, it could also enhance cell neurite growth, with a higher expression level of GAP-43 mRNA and protein. Conclusion “Tonifying qi and strengthening spleen” and “eliminating phlegm and strengthening intelligence” prescriptions from Shenghui soup have a positive effect on neurite growth. Their effects are related to the up-regulating expression of GAP-43. Electronic supplementary material The online version of this article (doi:10.1186/s12906-016-1339-y) contains supplementary material, which is available to authorized users.
Collapse
|
32
|
Wang Y, Li WY, Sun P, Jin ZS, Liu GB, Deng LX, Guan LX. Sciatic nerve regeneration in KLF7-transfected acellular nerve allografts. Neurol Res 2016; 38:242-54. [DOI: 10.1080/01616412.2015.1105584] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
33
|
Mehta ST, Luo X, Park KK, Bixby JL, Lemmon VP. Hyperactivated Stat3 boosts axon regeneration in the CNS. Exp Neurol 2016; 280:115-20. [PMID: 27060489 DOI: 10.1016/j.expneurol.2016.03.004] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Revised: 02/16/2016] [Accepted: 03/02/2016] [Indexed: 10/22/2022]
Abstract
Axonal regeneration after spinal cord injury (SCI) is intrinsically and extrinsically inhibited by multiple factors. One major factor contributing to intrinsic regeneration failure is the inability of mature neurons in the central nervous system (CNS) to activate regeneration-associated transcription factors (TFs) post-injury. A prior study identified TFs overexpressed in neurons of the peripheral nervous system (PNS) compared to the CNS; some of these could be involved in the ability of PNS neurons to regenerate. Of these, signal transducer and activator of transcription 3 (STAT3), as well its downstream regeneration-associated targets, showed a significant upregulation in PNS neurons relative to CNS neurons, and a constitutively active variant of Stat3 (Stat3CA) promoted neurite growth when expressed in cerebellar neurons (Lerch et al., 2012; Smith et al., 2011). To further enhance STAT3's neurite outgrowth enhancing activity, Stat3CA was fused with a viral activation domain (VP16). VP16 hyperactivates TFs by recruiting transcriptional co-factors to the DNA binding domain (Hirai et al., 2010). Overexpression of this VP16-Stat3CA chimera in primary cortical neurons led to a significant increase of neurite outgrowth as well as Stat3 transcriptional activity in vitro. Furthermore, in vivo transduction of retinal ganglion cells (RGCs) with AAV constructs expressing VP16-Stat3CA resulted in regeneration of optic nerve axons after injury, to a greater degree than for those expressing Stat3CA alone. These findings confirm and extend the concept that overexpression of hyperactivated transcription factors identified as functioning in PNS regeneration can promote axon regeneration in the CNS.
Collapse
Affiliation(s)
- Saloni T Mehta
- Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - Xueting Luo
- Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Department of Ophthalmology, Shanghai First People's Hospital, Shanghai Jiao Tong University School of Medicine; Shanghai, 200080, China.
| | - Kevin K Park
- Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - John L Bixby
- Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Center for Computational Science, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Molecular & Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - Vance P Lemmon
- Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Center for Computational Science, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| |
Collapse
|
34
|
Al-Ali H, Beckerman SR, Bixby JL, Lemmon VP. In vitro models of axon regeneration. Exp Neurol 2016; 287:423-434. [PMID: 26826447 DOI: 10.1016/j.expneurol.2016.01.020] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Revised: 01/20/2016] [Accepted: 01/25/2016] [Indexed: 12/31/2022]
Abstract
A variety of in vitro models have been developed to understand the mechanisms underlying the regenerative failure of central nervous system (CNS) axons, and to guide pre-clinical development of regeneration-promoting therapeutics. These range from single-cell based assays that typically focus on molecular mechanisms to organotypic assays that aim to recapitulate in vivo behavior. By utilizing a combination of models, researchers can balance the speed, convenience, and mechanistic resolution of simpler models with the biological relevance of more complex models. This review will discuss a number of models that have been used to build our understanding of the molecular mechanisms of CNS axon regeneration.
Collapse
Affiliation(s)
- Hassan Al-Ali
- Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Samuel R Beckerman
- Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - John L Bixby
- Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Center for Computational Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Molecular & Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - Vance P Lemmon
- Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Center for Computational Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| |
Collapse
|
35
|
DeFrancesco-Lisowitz A, Lindborg JA, Niemi JP, Zigmond RE. The neuroimmunology of degeneration and regeneration in the peripheral nervous system. Neuroscience 2015; 302:174-203. [PMID: 25242643 PMCID: PMC4366367 DOI: 10.1016/j.neuroscience.2014.09.027] [Citation(s) in RCA: 125] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Revised: 09/08/2014] [Accepted: 09/10/2014] [Indexed: 12/25/2022]
Abstract
Peripheral nerves regenerate following injury due to the effective activation of the intrinsic growth capacity of the neurons and the formation of a permissive pathway for outgrowth due to Wallerian degeneration (WD). WD and subsequent regeneration are significantly influenced by various immune cells and the cytokines they secrete. Although macrophages have long been known to play a vital role in the degenerative process, recent work has pointed to their importance in influencing the regenerative capacity of peripheral neurons. In this review, we focus on the various immune cells, cytokines, and chemokines that make regeneration possible in the peripheral nervous system, with specific attention placed on the role macrophages play in this process.
Collapse
Affiliation(s)
| | - J A Lindborg
- Department of Neurosciences, Case Western Reserve University, Cleveland OH 44106-4975
| | - J P Niemi
- Department of Neurosciences, Case Western Reserve University, Cleveland OH 44106-4975
| | - R E Zigmond
- Department of Neurosciences, Case Western Reserve University, Cleveland OH 44106-4975
| |
Collapse
|
36
|
Simpson MT, Venkatesh I, Callif BL, Thiel LK, Coley DM, Winsor KN, Wang Z, Kramer AA, Lerch JK, Blackmore MG. The tumor suppressor HHEX inhibits axon growth when prematurely expressed in developing central nervous system neurons. Mol Cell Neurosci 2015; 68:272-83. [PMID: 26306672 DOI: 10.1016/j.mcn.2015.08.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Revised: 07/30/2015] [Accepted: 08/17/2015] [Indexed: 01/21/2023] Open
Abstract
Neurons in the embryonic and peripheral nervous system respond to injury by activating transcriptional programs supportive of axon growth, ultimately resulting in functional recovery. In contrast, neurons in the adult central nervous system (CNS) possess a limited capacity to regenerate axons after injury, fundamentally constraining repair. Activating pro-regenerative gene expression in CNS neurons is a promising therapeutic approach, but progress is hampered by incomplete knowledge of the relevant transcription factors. An emerging hypothesis is that factors implicated in cellular growth and motility outside the nervous system may also control axon growth in neurons. We therefore tested sixty-nine transcription factors, previously identified as possessing tumor suppressive or oncogenic properties in non-neuronal cells, in assays of neurite outgrowth. This screen identified YAP1 and E2F1 as enhancers of neurite outgrowth, and PITX1, RBM14, ZBTB16, and HHEX as inhibitors. Follow-up experiments are focused on the tumor suppressor HHEX, one of the strongest growth inhibitors. HHEX is widely expressed in adult CNS neurons, including corticospinal tract neurons after spinal injury, but is present only in trace amounts in immature cortical neurons and adult peripheral neurons. HHEX overexpression in early postnatal cortical neurons reduced both initial axonogenesis and the rate of axon elongation, and domain deletion analysis strongly implicated transcriptional repression as the underlying mechanism. These findings suggest a role for HHEX in restricting axon growth in the developing CNS, and substantiate the hypothesis that previously identified oncogenes and tumor suppressors can play conserved roles in axon extension.
Collapse
Affiliation(s)
- Matthew T Simpson
- Marquette University, Department of Biomedical Sciences, 53201, United States
| | - Ishwariya Venkatesh
- Marquette University, Department of Biomedical Sciences, 53201, United States
| | - Ben L Callif
- Marquette University, Department of Biomedical Sciences, 53201, United States
| | - Laura K Thiel
- Marquette University, Department of Biomedical Sciences, 53201, United States
| | - Denise M Coley
- Marquette University, Department of Biomedical Sciences, 53201, United States
| | - Kristen N Winsor
- Marquette University, Department of Biomedical Sciences, 53201, United States
| | - Zimei Wang
- Marquette University, Department of Biomedical Sciences, 53201, United States
| | - Audra A Kramer
- Marquette University, Department of Biomedical Sciences, 53201, United States
| | - Jessica K Lerch
- The Ohio State University, The Center for Brain and Spinal Cord Repair, The Department of Neuroscience, 43210, United States
| | - Murray G Blackmore
- Marquette University, Department of Biomedical Sciences, 53201, United States.
| |
Collapse
|
37
|
Carmel JB, Young W, Hart RP. Flipping the transcriptional switch from myelin inhibition to axon growth in the CNS. Front Mol Neurosci 2015; 8:34. [PMID: 26236189 PMCID: PMC4505142 DOI: 10.3389/fnmol.2015.00034] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 07/06/2015] [Indexed: 01/01/2023] Open
Abstract
Poor regeneration of severed axons in the central nervous system (CNS) limits functional recovery. Regeneration failure involves interplay of inhibitory environmental elements and the growth state of the neuron. To find internal changes in gene expression that might overcome inhibitory environmental cues, we compared several paradigms that allow growth in the inhibitory environment. Conditions that allow axon growth by axotomized and cultured dorsal root ganglion (DRG) neurons on CNS myelin include immaturity (the first few postnatal days), high levels of cyclic adenosine mono phosphate (cAMP), and conditioning with a peripheral nerve lesion before explant. This shift from inhibition to growth depends on transcription. Seeking to understand the transcriptome changes that allow axon growth in the CNS, we collaborated with the Marie Filbin laboratory to identify several mRNAs that are functionally relevant, as determined by gain- and loss-of-function studies. In this Perspective, we review evidence from these experiments and discuss the merits of comparing multiple regenerative paradigms to identify a core transcriptional program for CNS axon regeneration.
Collapse
Affiliation(s)
- Jason B Carmel
- Brain Mind Research Institute and Departments of Pediatrics and Neurology, Weill Cornell Medical College New York, NY, USA ; Burke-Cornell Medical Research Institute White Plains, NY, USA
| | - Wise Young
- W.M. Keck Center for Collaborative Neuroscience and the Department of Cell Biology and Neuroscience, Rutgers University Piscataway, NJ, USA
| | - Ronald P Hart
- W.M. Keck Center for Collaborative Neuroscience and the Department of Cell Biology and Neuroscience, Rutgers University Piscataway, NJ, USA
| |
Collapse
|
38
|
Bermúdez M, Imaz-Rosshandler I, Rangel-Escareño C, Zeichner-David M, Arzate H, Mercado-Celis GE. CEMP1 Induces Transformation in Human Gingival Fibroblasts. PLoS One 2015; 10:e0127286. [PMID: 26011628 PMCID: PMC4444236 DOI: 10.1371/journal.pone.0127286] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 04/13/2015] [Indexed: 11/18/2022] Open
Abstract
Cementum Protein 1 (CEMP1) is a key regulator of cementogenesis. CEMP1 promotes cell attachment, differentiation, deposition rate, composition, and morphology of hydroxyapatite crystals formed by human cementoblastic cells. Its expression is restricted to cementoblasts and progenitor cell subpopulations present in the periodontal ligament. CEMP1 transfection into non-osteogenic cells such as adult human gingival fibroblasts results in differentiation of these cells into a “mineralizing” cell phenotype. Other studies have shown evidence that CEMP1 could have a therapeutic potential for the treatment of bone defects and regeneration of other mineralized tissues. To better understand CEMP1’s biological effects in vitro we investigated the consequences of its expression in human gingival fibroblasts (HGF) growing in non-mineralizing media by comparing gene expression profiles. We identified several mRNAs whose expression is modified by CEMP1 induction in HGF cells. Enrichment analysis showed that several of these newly expressed genes are involved in oncogenesis. Our results suggest that CEMP1 causes the transformation of HGF and NIH3T3 cells. CEMP1 is overexpressed in cancer cell lines. We also determined that the region spanning the CEMP1 locus is commonly amplified in a variety of cancers, and finally we found significant overexpression of CEMP1 in leukemia, cervix, breast, prostate and lung cancer. Our findings suggest that CEMP1 exerts modulation of a number of cellular genes, cellular development, cellular growth, cell death, and cell cycle, and molecules associated with cancer.
Collapse
Affiliation(s)
- Mercedes Bermúdez
- Laboratorio de Biología Periodontal, Facultad de Odontología, Universidad Nacional Autónoma de México, Mexico City, México
| | | | | | - Margarita Zeichner-David
- Ostrow School of Dentistry, University of Southern California, Los Angeles, California, United States of America
| | - Higinio Arzate
- Laboratorio de Biología Periodontal, Facultad de Odontología, Universidad Nacional Autónoma de México, Mexico City, México
| | | |
Collapse
|