1
|
Fang S, Ji Y, Shen Y, Yang S, Zhang H, Xin W, Shi W, Chen W. TET3 Contributes to Exercise-Induced Functional Axon Regeneration and Visual Restoration. Adv Biol (Weinh) 2024:e2400145. [PMID: 39007414 DOI: 10.1002/adbi.202400145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 05/27/2024] [Indexed: 07/16/2024]
Abstract
Axons have intrinsically poor regenerative capacity in the mature central nervous system (CNS), leading to permanent neurological impairments in individuals. There is growing evidence that exercise is a powerful physiological intervention that can obviously enhance cell rejuvenate capacity, but its molecular mechanisms that mediate the axonal regenerative benefits remain largely unclear. Using the eye as the CNS model, here it is first indicated that placing mice in an exercise stimulation environment induced DNA methylation patterns and transcriptomes of retinal ganglion cell, promoted axon regeneration after injury, and reversed vision loss in aged mice. These beneficial effects are dependent on the DNA demethylases TET3-mediated epigenetic effects, which increased the expression of genes associated with the regenerative growth programs, such as STAT3, Wnt5a, Klf6. Exercise training also shows with the improved mitochondrial and metabolic dysfunction in retinas and optic nerves via TET3. Collectively, these results suggested that the increased regenerative capacity induced by enhancing physical activity is mediated through epigenetic reprogramming in mouse model of optic nerve injury and in aged mouse. Understanding the molecular mechanism underlying exercise-dependent neuronal plasticity led to the identification of novel targets for ameliorating pathologies associated with etiologically diverse diseases.
Collapse
Affiliation(s)
- Si Fang
- Multiscale Research Institute of Complex Systems, Department of Integrative Oncology in Fudan University Shanghai Cancer Center, Jingan District Central Hospital of Shanghai, Department of Otorhinolaryngology-Head and Neck Surgery in Huashan Hospital, Fudan University, Shanghai, 200433, China
| | - Yunxiang Ji
- Multiscale Research Institute of Complex Systems, Department of Integrative Oncology in Fudan University Shanghai Cancer Center, Jingan District Central Hospital of Shanghai, Department of Otorhinolaryngology-Head and Neck Surgery in Huashan Hospital, Fudan University, Shanghai, 200433, China
| | - Yilan Shen
- Department of Nephrology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Simin Yang
- Multiscale Research Institute of Complex Systems, Department of Integrative Oncology in Fudan University Shanghai Cancer Center, Jingan District Central Hospital of Shanghai, Department of Otorhinolaryngology-Head and Neck Surgery in Huashan Hospital, Fudan University, Shanghai, 200433, China
- Department of Pharmacology, Basic Medical College, Anhui Medical University, Hefei, 230032, China
| | - Hongli Zhang
- Multiscale Research Institute of Complex Systems, Department of Integrative Oncology in Fudan University Shanghai Cancer Center, Jingan District Central Hospital of Shanghai, Department of Otorhinolaryngology-Head and Neck Surgery in Huashan Hospital, Fudan University, Shanghai, 200433, China
- Department of Nephrology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Wenfeng Xin
- College of Notoginseng Medicine and Pharmacy, Wenshan University, Wenshan, 663000, China
| | - Weidong Shi
- Multiscale Research Institute of Complex Systems, Department of Integrative Oncology in Fudan University Shanghai Cancer Center, Jingan District Central Hospital of Shanghai, Department of Otorhinolaryngology-Head and Neck Surgery in Huashan Hospital, Fudan University, Shanghai, 200433, China
| | - Wei Chen
- Multiscale Research Institute of Complex Systems, Department of Integrative Oncology in Fudan University Shanghai Cancer Center, Jingan District Central Hospital of Shanghai, Department of Otorhinolaryngology-Head and Neck Surgery in Huashan Hospital, Fudan University, Shanghai, 200433, China
| |
Collapse
|
2
|
Wang Y, Yang B, Huang Z, Yang Z, Wang J, Ao Q, Yin G, Li Y. Progress and mechanism of graphene oxide-composited materials in application of peripheral nerve repair. Colloids Surf B Biointerfaces 2024; 234:113672. [PMID: 38071946 DOI: 10.1016/j.colsurfb.2023.113672] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 11/21/2023] [Accepted: 11/23/2023] [Indexed: 02/09/2024]
Abstract
Peripheral nerve injuries (PNI) are one of the most common nerve injuries, and graphene oxide (GO) has demonstrated significant potential in the treatment of PNI. GO could enhance the proliferation, adhesion, migration, and differentiation of neuronal cells by upregulating the expression of relevant proteins, and regulate the angiogenesis process and immune response. Therefore, GO is a suitable additional component for fabricating artificial nerve scaffolds (ANS), in which the slight addition of GO could improve the physicochemical performance of the matrix materials, through hydrogen bonds and electrostatic attraction. GO-composited ANS can increase the expression of nerve regeneration-associated genes and factors, promoting angiogenesis by activating the RAS/MAPK and AKT-eNOS-VEGF signaling pathway, respectively. Moreover, GO could be metabolized and excreted from the body through the pathway of peroxidase degradation in vivo. Consequently, the application of GO in PNI regeneration exhibits significant potential for transitioning from laboratory research to clinical use.
Collapse
Affiliation(s)
- Yulin Wang
- College of Biomedical Engineering, Sichuan University, China; Institute of Regulatory Science for Medical Devices, Sichuan University, China
| | - Bing Yang
- College of Biomedical Engineering, Sichuan University, China; Precision Medical Center of Southwest China Hospital, Sichuan University, China
| | - Zhongbing Huang
- College of Biomedical Engineering, Sichuan University, China.
| | - Zhaopu Yang
- Center for Drug Inspection, Guizhou Medical Products Administration, China
| | - Juan Wang
- College of Biomedical Engineering, Sichuan University, China
| | - Qiang Ao
- College of Biomedical Engineering, Sichuan University, China; Institute of Regulatory Science for Medical Devices, Sichuan University, China
| | - Guangfu Yin
- College of Biomedical Engineering, Sichuan University, China
| | - Ya Li
- College of Biomedical Engineering, Sichuan University, China; Institute of Regulatory Science for Medical Devices, Sichuan University, China
| |
Collapse
|
3
|
Mcloughlin KJ, Aladdad AM, Payne AJ, Boda AI, Nieto-Gomez S, Kador KE. Purification of retinal ganglion cells using low-pressure flow cytometry. Front Mol Neurosci 2023; 16:1149024. [PMID: 37547921 PMCID: PMC10400357 DOI: 10.3389/fnmol.2023.1149024] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 07/05/2023] [Indexed: 08/08/2023] Open
Abstract
Purified Retinal Ganglion Cells (RGCs) for in vitro study have been a valuable tool in the study of neural regeneration and in the development of therapies to treat glaucoma. Traditionally, RGCs have been isolated from early postnatal rats and mice, and more recently from human in vitro derived retinal organoids using a two-step immunopanning technique based upon the expression of Thy-1. This technique, however, limits the time periods from which RGCs can be isolated, missing the earliest born RGCs at which time the greatest stage of axon growth occurs, as well as being limited in its use with models of retinal degeneration as Thy-1 is downregulated following injury. While fluorescence associated cell sorting (FACS) in combination with new optogenetically labeled RGCs would be able to overcome this limitation, the use of traditional FACS sorters has been limited to genomic and proteomic studies, as RGCs have little to no survival post-sorting. Here we describe a new method for RGC isolation utilizing a combined immunopanning-fluorescence associated cell sorting (IP-FACS) protocol that initially depletes macrophages and photoreceptors, using immunopanning to enrich for RGCs before using low-pressure FACS to isolate these cells. We demonstrate that RGCs isolated via IP-FACS when compared to RGCs isolated via immunopanning at the same age have similar purity as measured by antibody staining and qRT-PCR; survival as measured by live dead staining; neurite outgrowth; and electrophysiological properties as measured by calcium release response to glutamate. Finally, we demonstrate the ability to isolate RGCs from early embryonic mice prior to the expression of Thy-1 using Brn3b-eGFP optogenetically labeled cells. This method provides a new approach for the isolation of RGCs for the study of early developed RGCs, the study of RGC subtypes and the isolation of RGCs for cell transplantation studies.
Collapse
Affiliation(s)
- Kiran J. Mcloughlin
- Department of Ophthalmology, School of Medicine, University of Missouri-Kansas City, Kansas City, MO, United States
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City, Kansas City, MO, United States
| | - Afnan M. Aladdad
- Department of Ophthalmology, School of Medicine, University of Missouri-Kansas City, Kansas City, MO, United States
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City, Kansas City, MO, United States
| | - Andrew J. Payne
- Department of Ophthalmology, School of Medicine, University of Missouri-Kansas City, Kansas City, MO, United States
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City, Kansas City, MO, United States
| | - Anna I. Boda
- Department of Ophthalmology, School of Medicine, University of Missouri-Kansas City, Kansas City, MO, United States
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City, Kansas City, MO, United States
| | - Sayra Nieto-Gomez
- Department of Ophthalmology, School of Medicine, University of Missouri-Kansas City, Kansas City, MO, United States
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City, Kansas City, MO, United States
| | - Karl E. Kador
- Department of Ophthalmology, School of Medicine, University of Missouri-Kansas City, Kansas City, MO, United States
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City, Kansas City, MO, United States
| |
Collapse
|
4
|
Ghose A, Pullarkat P. The role of mechanics in axonal stability and development. Semin Cell Dev Biol 2023; 140:22-34. [PMID: 35786351 PMCID: PMC7615100 DOI: 10.1016/j.semcdb.2022.06.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 06/05/2022] [Accepted: 06/13/2022] [Indexed: 01/28/2023]
Abstract
Much of the focus of neuronal cell biology has been devoted to growth cone guidance, synaptogenesis, synaptic activity, plasticity, etc. The axonal shaft too has received much attention, mainly for its astounding ability to transmit action potentials and the transport of material over long distances. For these functions, the axonal cytoskeleton and membrane have been often assumed to play static structural roles. Recent experiments have changed this view by revealing an ultrastructure much richer in features than previously perceived and one that seems to be maintained at a dynamic steady state. The role of mechanics in this is only beginning to be broadly appreciated and appears to involve passive and active modes of coupling different biopolymer filaments, filament turnover dynamics and membrane biophysics. Axons, being unique cellular processes in terms of high aspect ratios and often extreme lengths, also exhibit unique passive mechanical properties that might have evolved to stabilize them under mechanical stress. In this review, we summarize the experiments that have exposed some of these features. It is our view that axonal mechanics deserves much more attention not only due to its significance in the development and maintenance of the nervous system but also due to the susceptibility of axons to injury and neurodegeneration.
Collapse
Affiliation(s)
- Aurnab Ghose
- Indian Institute of Science Education and Research, Pune 411 008, India.
| | - Pramod Pullarkat
- Raman Research Institute, C. V. Raman Avenue, Bengaluru 560 080, India.
| |
Collapse
|
5
|
Raffa V. Force: A messenger of axon outgrowth. Semin Cell Dev Biol 2023; 140:3-12. [PMID: 35817654 DOI: 10.1016/j.semcdb.2022.07.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 07/05/2022] [Accepted: 07/05/2022] [Indexed: 01/28/2023]
Abstract
The axon is a sophisticated macromolecular machine composed of interrelated parts that transmit signals like spur gears transfer motion between parallel shafts. The growth cone is a fine sensor that integrates mechanical and chemical cues and transduces these signals through the generation of a traction force that pushes the tip and pulls the axon shaft forward. The axon shaft, in turn, senses this pulling force and transduces this signal in an orchestrated response, coordinating cytoskeleton remodeling and intercalated mass addition to sustain and support the advancing of the tip. Extensive research suggests that the direct application of active force is per se a powerful inducer of axon growth, potentially bypassing the contribution of the growth cone. This review provides a critical perspective on current knowledge of how the force is a messenger of axon growth and its mode of action for controlling navigation, including aspects that remain unclear. It also focuses on novel approaches and tools designed to mechanically manipulate axons, and discusses their implications in terms of potential novel therapies for re-wiring the nervous system.
Collapse
Affiliation(s)
- Vittoria Raffa
- Department of Biology, University of Pisa, SS12 Abetone e Brennero, 4, 56127 Pisa, Italy.
| |
Collapse
|
6
|
Falconieri A, De Vincentiis S, Cappello V, Convertino D, Das R, Ghignoli S, Figoli S, Luin S, Català-Castro F, Marchetti L, Borello U, Krieg M, Raffa V. Axonal plasticity in response to active forces generated through magnetic nano-pulling. Cell Rep 2022; 42:111912. [PMID: 36640304 PMCID: PMC9902337 DOI: 10.1016/j.celrep.2022.111912] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 11/16/2022] [Accepted: 12/12/2022] [Indexed: 12/30/2022] Open
Abstract
Mechanical force is crucial in guiding axon outgrowth before and after synapse formation. This process is referred to as "stretch growth." However, how neurons transduce mechanical input into signaling pathways remains poorly understood. Another open question is how stretch growth is coupled in time with the intercalated addition of new mass along the entire axon. Here, we demonstrate that active mechanical force generated by magnetic nano-pulling induces remodeling of the axonal cytoskeleton. Specifically, the increase in the axonal density of microtubules induced by nano-pulling leads to an accumulation of organelles and signaling vesicles, which, in turn, promotes local translation by increasing the probability of assembly of the "translation factories." Modulation of axonal transport and local translation sustains enhanced axon outgrowth and synapse maturation.
Collapse
Affiliation(s)
| | - Sara De Vincentiis
- Department of Biology, Università di Pisa, 56127 Pisa, Italy,The Barcelona Institute of Science and Technology, Institut de Ciències Fotòniques, ICFO, 08860 Castelldefels, Spain
| | - Valentina Cappello
- Center for Materials Interfaces, Istituto Italiano di Tecnologia, 56025 Pontedera, Italy
| | - Domenica Convertino
- Center for Nanotechnology Innovation @NEST, Istituto Italiano di Tecnologia, 56127 Pisa, Italy
| | - Ravi Das
- The Barcelona Institute of Science and Technology, Institut de Ciències Fotòniques, ICFO, 08860 Castelldefels, Spain
| | | | - Sofia Figoli
- Department of Biology, Università di Pisa, 56127 Pisa, Italy
| | - Stefano Luin
- National Enterprise for NanoScience and NanoTechnology (NEST) Laboratory, Scuola Normale Superiore, 56127 Pisa, Italy
| | - Frederic Català-Castro
- The Barcelona Institute of Science and Technology, Institut de Ciències Fotòniques, ICFO, 08860 Castelldefels, Spain
| | - Laura Marchetti
- Center for Nanotechnology Innovation @NEST, Istituto Italiano di Tecnologia, 56127 Pisa, Italy,Department of Pharmacy, Università di Pisa, 56126 Pisa, Italy
| | - Ugo Borello
- Department of Biology, Università di Pisa, 56127 Pisa, Italy
| | - Michael Krieg
- The Barcelona Institute of Science and Technology, Institut de Ciències Fotòniques, ICFO, 08860 Castelldefels, Spain
| | - Vittoria Raffa
- Department of Biology, Università di Pisa, 56127 Pisa, Italy.
| |
Collapse
|
7
|
Sun Q, Xu W, Piao J, Su J, Ge T, Cui R, Yang W, Li B. Transcription factors are potential therapeutic targets in epilepsy. J Cell Mol Med 2022; 26:4875-4885. [PMID: 36065764 PMCID: PMC9549512 DOI: 10.1111/jcmm.17518] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 06/28/2022] [Accepted: 07/01/2022] [Indexed: 11/29/2022] Open
Abstract
Academics generally believe that imbalance between excitation and inhibition of the nervous system is the root cause of epilepsy. However, the aetiology of epilepsy is complex, and its pathogenesis remains unclear. Many studies have shown that epilepsy is closely related to genetic factors. Additionally, the involvement of a variety of tumour‐related transcription factors in the pathogenesis of epilepsy has been confirmed, which also confirms the heredity of epilepsy. In this review, we summarize the existing research on a variety of transcription factors and epilepsy and present relevant evidence related to transcription factors that may be targets in epilepsy. This information is of great significance for revealing the in‐depth molecular and cellular mechanisms of epilepsy.
Collapse
Affiliation(s)
- Qihan Sun
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Wenbo Xu
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Jingjing Piao
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Jingyun Su
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Tongtong Ge
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Ranji Cui
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Wei Yang
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Bingjin Li
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
8
|
Yin C, Ji Y, Ma N, Chen K, Zhang W, Bai D, Jia X, Xia S, Yin H. RNA-seq analysis reveals potential molecular mechanisms of ZNF580/ZFP580 promoting neuronal survival and inhibiting apoptosis after Hypoxic-ischemic brain damage. Neuroscience 2021; 483:52-65. [PMID: 34929337 DOI: 10.1016/j.neuroscience.2021.12.018] [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: 10/27/2021] [Revised: 12/09/2021] [Accepted: 12/12/2021] [Indexed: 10/19/2022]
Abstract
Neonatal hypoxic-ischemic brain damage (HIBD) is one of the main causes of neonatal acute death and chronic nervous system impairment, but still lacks effective treatments. ZNF580/ZFP580, reported in our previous studies, may be a newly identified member of the Krüppel-like factor (KLF) family, and has anti-apoptotic effects during ischemic myocardial injury. In the present study, we showed that the expression levels of both ZFP580/ZNF580 mRNA and protein increased significantly in neonatal HIBD rats and oxygen-glucose deprivation (OGD) SH-SY5Y cell models. ZNF580 overexpression promoted neuron survival and suppressed neuron apoptosis after OGD in neuron-like SH-SY5Y cells, while interference with ZNF580 resulted in the opposite results. RNA-seq analysis identified 248 differentially-expressed genes (DEGs) between ZNF580 overexpression SH-SY5Y cells and interference-expressed SH-SY5Y cells. Gene Ontology functional enrichment analysis showed that these DEGs played significant roles in the growth, development, and regeneration of axons, DNA biosynthetic processes, DNA replication, and apoptosis. Kyoto Encyclopedia of Genes and Genomes enrichment analysis indicated that these DEGs were found in some pathways, including ferroptosis, glutamatergic synapses, protein processing in the endoplasmic reticulum, estrogen signaling pathways, the TGF-beta signaling pathway, and the longevity regulating pathway. The qRT-PCR validation results were consistent with RNA-seq results, which showed that HSPA5, IGFBP3, NTN4, and KLF9 increased in ZNF580-overexpressed SH-SY5Y cells and decreased in interference-expressed SH-SY5Y cells, when compared with normal cells. Together, the results suggested that ZNF580 targeted these genes to inhibit neuronal apoptosis.
Collapse
Affiliation(s)
- Chongjuan Yin
- First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China; Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yansu Ji
- Characteristic Medical Center of Chinese People's Armed Police Force, Tianjin, Hebei, China
| | - Ning Ma
- Shanxi Medical University, Taiyuan, Shanxi, China
| | - Kai Chen
- Characteristic Medical Center of Chinese People's Armed Police Force, Tianjin, Hebei, China
| | - Wencheng Zhang
- Characteristic Medical Center of Chinese People's Armed Police Force, Tianjin, Hebei, China
| | - Dan Bai
- First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xiaojun Jia
- First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Shihai Xia
- Characteristic Medical Center of Chinese People's Armed Police Force, Tianjin, Hebei, China.
| | - Huaiqing Yin
- First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China.
| |
Collapse
|
9
|
Fibroblast Growth Factor 13 Facilitates Peripheral Nerve Regeneration through Maintaining Microtubule Stability. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5481228. [PMID: 34457114 PMCID: PMC8397546 DOI: 10.1155/2021/5481228] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 08/02/2021] [Indexed: 12/19/2022]
Abstract
Peripheral nerve injury (PNI), resulting in the impairment of myelin sheaths and axons, seriously affects the transmission of sensory or motor nerves. Growth factors (GFs) provide a biological microenvironment for supporting nerve regrowth and have become a promising alternative for repairing PNI. As one number of intracellular growth factor family, fibroblast growth factor 13 (FGF13) was regard as a microtubule-stabilizing protein for regulating cytoskeletal plasticity and neuronal polarization. However, the therapeutic efficiency and underlying mechanism of FGF13 for treating PNI remained unknown. Here, the application of lentivirus that overexpressed FGF13 was delivered directly to the lesion site of transverse sciatic nerve for promoting peripheral nerve regeneration. Through behavioral analysis and histological and ultrastructure examinations, we found that FGF13 not only facilitated motor and sense functional recovery but also enhanced axon elongation and remyelination. Furthermore, pretreatment with FGF13 also promoted Schwann cell (SC) viability and upregulated the expression cellular microtubule-associated proteins in vitro PNI model. These data indicated FGF13 therapeutic effect was closely related to maintain cellular microtubule stability. Thus, this work provides the evident that FGF13-medicated microtubule stability is necessary for promoting peripheral nerve repair following PNI, highlighting the potential therapeutic value of FGF13 on ameliorating injured nerve recovery.
Collapse
|
10
|
MicroRNA-7 promotes motor function recovery following spinal cord injury in mice. Biochem Biophys Res Commun 2021; 573:80-85. [PMID: 34399097 DOI: 10.1016/j.bbrc.2021.08.020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 08/05/2021] [Indexed: 01/04/2023]
Abstract
Spinal cord injury (SCI) is a devastating neurological condition for which there are no effective therapies. Following an initial injury, there is a cascade of multiple downstream events termed secondary injury. Thus, therapeutic approaches targeting a single pathway may not offer the best solution for treating SCI. One of the most attractive properties of microRNAs (miR) as potential therapeutics is that they are highly effective in regulating complex biological pathways by targeting multiple genes and pathways. The current study investigated the role of miR-7-5p (miR-7), which was previously shown to have neuroprotective functions, in promoting motor function recovery following SCI. We used an adeno-associated virus 1 (AAV1) vector to deliver the gene encoding miR-7 to the spinal cord of adult mice and found that this virus was mainly transduced into the neurons of the spinal cord. Transduction of AAV1-miR-7 improved hindlimb locomotor function following SCI over an 8-week observation period. This improvement was accompanied by reduced neuronal loss in the lesion. In addition, the beneficial effect of miR-7 was associated with enhanced levels of TH-positive axons in the lesion. Taken together, we suggest that miR-7 improves motor function recovery after SCI by protecting neuronal death and increasing axon levels. These findings suggest that miR-7 could be developed as a potential treatment for SCI in human.
Collapse
|
11
|
Pita-Thomas W, Gonçalves TM, Kumar A, Zhao G, Cavalli V. Genome-wide chromatin accessibility analyses provide a map for enhancing optic nerve regeneration. Sci Rep 2021; 11:14924. [PMID: 34290335 PMCID: PMC8295311 DOI: 10.1038/s41598-021-94341-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 07/05/2021] [Indexed: 11/21/2022] Open
Abstract
Retinal Ganglion Cells (RGCs) lose their ability to grow axons during development. Adult RGCs thus fail to regenerate their axons after injury, leading to vision loss. To uncover mechanisms that promote regeneration of RGC axons, we identified transcription factors (TF) and open chromatin regions that are enriched in rat embryonic RGCs (high axon growth capacity) compared to postnatal RGCs (low axon growth capacity). We found that developmental stage-specific gene expression changes correlated with changes in promoter chromatin accessibility. Binding motifs for TFs such as CREB, CTCF, JUN and YY1 were enriched in the regions of the chromatin that were more accessible in embryonic RGCs. Proteomic analysis of purified rat RGC nuclei confirmed the expression of TFs with potential role in axon growth such as CREB, CTCF, YY1, and JUND. The CREB/ATF binding motif was widespread at the open chromatin region of known pro-regenerative TFs, supporting a role of CREB in regulating axon regeneration. Consistently, overexpression of CREB fused to the VP64 transactivation domain in mouse RGCs promoted axon regeneration after optic nerve injury. Our study provides a map of the chromatin accessibility during RGC development and highlights that TF associated with developmental axon growth can stimulate axon regeneration in mature RGC.
Collapse
Affiliation(s)
- Wolfgang Pita-Thomas
- Department of Neuroscience, Washington University School of Medicine, St Louis, MO, 63110, USA
| | | | - Ajeet Kumar
- Department of Neuroscience, Washington University School of Medicine, St Louis, MO, 63110, USA
| | - Guoyan Zhao
- Department of Neuroscience, Washington University School of Medicine, St Louis, MO, 63110, USA. .,Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, 63110, USA.
| | - Valeria Cavalli
- Department of Neuroscience, Washington University School of Medicine, St Louis, MO, 63110, USA. .,Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, 63110, USA. .,Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| |
Collapse
|
12
|
Sitko AA, Goodrich LV. Making sense of neural development by comparing wiring strategies for seeing and hearing. Science 2021; 371:eaaz6317. [PMID: 33414193 PMCID: PMC8034811 DOI: 10.1126/science.aaz6317] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The ability to perceive and interact with the world depends on a diverse array of neural circuits specialized for carrying out specific computations. Each circuit is assembled using a relatively limited number of molecules and common developmental steps, from cell fate specification to activity-dependent synaptic refinement. Given this shared toolkit, how do individual circuits acquire their characteristic properties? We explore this question by comparing development of the circuitry for seeing and hearing, highlighting a few examples where differences in each system's sensory demands necessitate different developmental strategies.
Collapse
Affiliation(s)
- A A Sitko
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - L V Goodrich
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
13
|
Kampanis V, Tolou-Dabbaghian B, Zhou L, Roth W, Puttagunta R. Cyclic Stretch of Either PNS or CNS Located Nerves Can Stimulate Neurite Outgrowth. Cells 2020; 10:cells10010032. [PMID: 33379276 PMCID: PMC7824691 DOI: 10.3390/cells10010032] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 12/15/2022] Open
Abstract
The central nervous system (CNS) does not recover from traumatic axonal injury, but the peripheral nervous system (PNS) does. We hypothesize that this fundamental difference in regenerative capacity may be based upon the absence of stimulatory mechanical forces in the CNS due to the protective rigidity of the vertebral column and skull. We developed a bioreactor to apply low-strain cyclic axonal stretch to adult rat dorsal root ganglia (DRG) connected to either the peripheral or central nerves in an explant model for inducing axonal growth. In response, larger diameter DRG neurons, mechanoreceptors and proprioceptors showed enhanced neurite outgrowth as well as increased Activating Transcription Factor 3 (ATF3).
Collapse
Affiliation(s)
- Vasileios Kampanis
- Laboratory for Experimental Neuroregeneration, Spinal Cord Injury Center, Heidelberg University Hospital, 69118 Heidelberg, Germany; (V.K.); (B.T.-D.)
| | - Bahardokht Tolou-Dabbaghian
- Laboratory for Experimental Neuroregeneration, Spinal Cord Injury Center, Heidelberg University Hospital, 69118 Heidelberg, Germany; (V.K.); (B.T.-D.)
| | - Luming Zhou
- Laboratory of NeuroRegeneration and Repair, Hertie Institute for Clinical Brain Research, University of Tübingen, 72076 Tübingen, Germany;
| | - Wolfgang Roth
- Laboratory for Experimental Neurorehabilitation, Heidelberg University Hospital, 69118 Heidelberg, Germany;
| | - Radhika Puttagunta
- Laboratory for Experimental Neuroregeneration, Spinal Cord Injury Center, Heidelberg University Hospital, 69118 Heidelberg, Germany; (V.K.); (B.T.-D.)
- Correspondence:
| |
Collapse
|
14
|
Fundamentals and Current Strategies for Peripheral Nerve Repair and Regeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1249:173-201. [PMID: 32602098 DOI: 10.1007/978-981-15-3258-0_12] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
A body of evidence indicates that peripheral nerves have an extraordinary yet limited capacity to regenerate after an injury. Peripheral nerve injuries have confounded professionals in this field, from neuroscientists to neurologists, plastic surgeons, and the scientific community. Despite all the efforts, full functional recovery is still seldom. The inadequate results attained with the "gold standard" autograft procedure still encourage a dynamic and energetic research around the world for establishing good performing tissue-engineered alternative grafts. Resourcing to nerve guidance conduits, a variety of methods have been experimentally used to bridge peripheral nerve gaps of limited size, up to 30-40 mm in length, in humans. Herein, we aim to summarize the fundamentals related to peripheral nerve anatomy and overview the challenges and scientific evidences related to peripheral nerve injury and repair mechanisms. The most relevant reports dealing with the use of both synthetic and natural-based biomaterials used in tissue engineering strategies when treatment of nerve injuries is envisioned are also discussed in depth, along with the state-of-the-art approaches in this field.
Collapse
|
15
|
Manipulation of Axonal Outgrowth via Exogenous Low Forces. Int J Mol Sci 2020; 21:ijms21218009. [PMID: 33126477 PMCID: PMC7663625 DOI: 10.3390/ijms21218009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 10/21/2020] [Accepted: 10/23/2020] [Indexed: 12/24/2022] Open
Abstract
Neurons are mechanosensitive cells. The role of mechanical force in the process of neurite initiation, elongation and sprouting; nerve fasciculation; and neuron maturation continues to attract considerable interest among scientists. Force is an endogenous signal that stimulates all these processes in vivo. The axon is able to sense force, generate force and, ultimately, transduce the force in a signal for growth. This opens up fascinating scenarios. How are forces generated and sensed in vivo? Which molecular mechanisms are responsible for this mechanotransduction signal? Can we exploit exogenously applied forces to mimic and control this process? How can these extremely low forces be generated in vivo in a non-invasive manner? Can these methodologies for force generation be used in regenerative therapies? This review addresses these questions, providing a general overview of current knowledge on the applications of exogenous forces to manipulate axonal outgrowth, with a special focus on forces whose magnitude is similar to those generated in vivo. We also review the principal methodologies for applying these forces, providing new inspiration and insights into the potential of this approach for future regenerative therapies.
Collapse
|
16
|
Kogo Y, Seto C, Totani Y, Mochizuki M, Nakahara T, Oka K, Yoshioka T, Ito E. Rapid differentiation of human dental pulp stem cells to neuron-like cells by high K + stimulation. Biophys Physicobiol 2020; 17:132-139. [PMID: 33240740 PMCID: PMC7671740 DOI: 10.2142/biophysico.bsj-2020023] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 09/18/2020] [Indexed: 02/06/2023] Open
Abstract
As human-origin cells, human dental pulp stem cells (hDPSCs) are thought to be potentially useful for biological and medical experiments. They are easily obtained from lost primary teeth or extracted wisdom teeth, and they are mesenchymal stem cells that are known to differentiate into osteoblasts, chondrocytes, and adipocytes. Although hDPSCs originate from neural crest cells, it is difficult to induce hDPSCs to differentiate into neuron-like cells. To facilitate their differentiation into neuron-like cells, we evaluated various differentiation conditions. Activation of K+ channels is thought to regulate the intracellular Ca2+ concentration, allowing for manipulation of the cell cycle to induce the differentiation of hDPSCs. Therefore, in addition to a conventional neural cell differentiation protocol, we activated K+ channels in hDPSCs. Immunocyto-chemistry and real-time PCR revealed that applying a combination of 3 stimuli (high K+ solution, epigenetic reprogramming solution, and neural differentiation solution) to hDPSCs increased their expression of neuronal markers, such as β3-tubulin, postsynaptic density protein 95, and nestin within 5 days, which led to their rapid differentiation into neuron-like cells. Our findings indicate that epigenetic reprogramming along with cell cycle regulation by stimulation with high K+ accelerated the differentiation of hDPSCs into neuron-like cells. Therefore, hDPSCs can be used in various ways as neuron-like cells by manipulating their cell cycle.
Collapse
Affiliation(s)
- Yuki Kogo
- Department of Biology, Waseda University, Tokyo 162-8480, Japan
| | - Chiaki Seto
- Department of Biology, Waseda University, Tokyo 162-8480, Japan
| | - Yuki Totani
- Department of Biology, Waseda University, Tokyo 162-8480, Japan
| | - Mai Mochizuki
- Department of Life Science Dentistry, The Nippon Dental University, Tokyo 102-8159, Japan
- Department of Developmental and Regenerative Dentistry, The Nippon Dental University School of Life Dentistry at Tokyo, Tokyo 102-8159, Japan
- Department of Bioscience and Informatics, Faculty of Science and Technology, Keio University, Yokohama 223-8522, Japan
- Waseda Research Institute for Science and Engineering, Waseda University, Tokyo 169-8555, Japan
| | - Taka Nakahara
- Department of Developmental and Regenerative Dentistry, The Nippon Dental University School of Life Dentistry at Tokyo, Tokyo 102-8159, Japan
| | - Kotaro Oka
- Department of Bioscience and Informatics, Faculty of Science and Technology, Keio University, Yokohama 223-8522, Japan
- Waseda Research Institute for Science and Engineering, Waseda University, Tokyo 169-8555, Japan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Tohru Yoshioka
- Waseda Research Institute for Science and Engineering, Waseda University, Tokyo 169-8555, Japan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Etsuro Ito
- Department of Biology, Waseda University, Tokyo 162-8480, Japan
- Waseda Research Institute for Science and Engineering, Waseda University, Tokyo 169-8555, Japan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| |
Collapse
|
17
|
Neuroprotective Strategies for Retinal Ganglion Cell Degeneration: Current Status and Challenges Ahead. Int J Mol Sci 2020; 21:ijms21072262. [PMID: 32218163 PMCID: PMC7177277 DOI: 10.3390/ijms21072262] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 03/19/2020] [Accepted: 03/20/2020] [Indexed: 12/12/2022] Open
Abstract
The retinal ganglion cells (RGCs) are the output cells of the retina into the brain. In mammals, these cells are not able to regenerate their axons after optic nerve injury, leaving the patients with optic neuropathies with permanent visual loss. An effective RGCs-directed therapy could provide a beneficial effect to prevent the progression of the disease. Axonal injury leads to the functional loss of RGCs and subsequently induces neuronal death, and axonal regeneration would be essential to restore the neuronal connectivity, and to reestablish the function of the visual system. The manipulation of several intrinsic and extrinsic factors has been proposed in order to stimulate axonal regeneration and functional repairing of axonal connections in the visual pathway. However, there is a missing point in the process since, until now, there is no therapeutic strategy directed to promote axonal regeneration of RGCs as a therapeutic approach for optic neuropathies.
Collapse
|
18
|
Carvalho CR, Silva-Correia J, Oliveira JM, Reis RL. Nanotechnology in peripheral nerve repair and reconstruction. Adv Drug Deliv Rev 2019; 148:308-343. [PMID: 30639255 DOI: 10.1016/j.addr.2019.01.006] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 09/20/2018] [Accepted: 01/05/2019] [Indexed: 02/07/2023]
Affiliation(s)
- Cristiana R Carvalho
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal; The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, AvePark, 4805-017 Barco, Guimarães, Portugal
| | - Joana Silva-Correia
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Joaquim M Oliveira
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal; The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, AvePark, 4805-017 Barco, Guimarães, Portugal
| | - Rui L Reis
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal; The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, AvePark, 4805-017 Barco, Guimarães, Portugal.
| |
Collapse
|
19
|
Response of mechanically-created neurites to extension. J Mech Behav Biomed Mater 2019; 98:121-130. [PMID: 31229904 DOI: 10.1016/j.jmbbm.2019.06.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Revised: 05/24/2019] [Accepted: 06/17/2019] [Indexed: 11/22/2022]
Abstract
We use micromanipulation techniques and real-time particle tracking to develop an approach to study specific attributes of neuron mechanics. We use a mechanical probe composed of a hollow micropipette with its tip fixed to a functionalized bead to induce the formation of a neurite in a sample of rat hippocampal neurons. We then move the sample relative to the pipette tip, elongating the neurite while simultaneously measuring its tension by optically tracking the deflection of the beaded tip. By calibrating the spring constant of the pipette, we can convert this deflection to a force. We use this technique to obtain uniaxial strain measurements of induced neurites and investigate the dependence of the force-extension relationship on mechanical pull speed. We show that in the range of pull speeds studied (0.05-1.8 μm/s), the variation in the work to extend a neurite 10 μm is consistent across pull speeds. We do not observe statistically significant rate-dependent effects in the force-extension profiles; instead we find the same quadratic behaviour (with parameters drawn from the same distributions) at each pull speed.
Collapse
|
20
|
Padmanabhan P, Goodhill GJ. Axon growth regulation by a bistable molecular switch. Proc Biol Sci 2019; 285:rspb.2017.2618. [PMID: 29669897 DOI: 10.1098/rspb.2017.2618] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 03/19/2018] [Indexed: 02/07/2023] Open
Abstract
For the brain to function properly, its neurons must make the right connections during neural development. A key aspect of this process is the tight regulation of axon growth as axons navigate towards their targets. Neuronal growth cones at the tips of developing axons switch between growth and paused states during axonal pathfinding, and this switching behaviour determines the heterogeneous axon growth rates observed during brain development. The mechanisms controlling this switching behaviour, however, remain largely unknown. Here, using mathematical modelling, we predict that the molecular interaction network involved in axon growth can exhibit bistability, with one state representing a fast-growing growth cone state and the other a paused growth cone state. Owing to stochastic effects, even in an unchanging environment, model growth cones reversibly switch between growth and paused states. Our model further predicts that environmental signals could regulate axon growth rate by controlling the rates of switching between the two states. Our study presents a new conceptual understanding of growth cone switching behaviour, and suggests that axon guidance may be controlled by both cell-extrinsic factors and cell-intrinsic growth regulatory mechanisms.
Collapse
Affiliation(s)
- Pranesh Padmanabhan
- Queensland Brain Institute, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - Geoffrey J Goodhill
- Queensland Brain Institute, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia .,School of Mathematics and Physics, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| |
Collapse
|
21
|
Bagonis MM, Fusco L, Pertz O, Danuser G. Automated profiling of growth cone heterogeneity defines relations between morphology and motility. J Cell Biol 2019; 218:350-379. [PMID: 30523041 PMCID: PMC6314545 DOI: 10.1083/jcb.201711023] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 09/26/2018] [Accepted: 11/08/2018] [Indexed: 12/14/2022] Open
Abstract
Growth cones are complex, motile structures at the tip of an outgrowing neurite. They often exhibit a high density of filopodia (thin actin bundles), which complicates the unbiased quantification of their morphologies by software. Contemporary image processing methods require extensive tuning of segmentation parameters, require significant manual curation, and are often not sufficiently adaptable to capture morphology changes associated with switches in regulatory signals. To overcome these limitations, we developed Growth Cone Analyzer (GCA). GCA is designed to quantify growth cone morphodynamics from time-lapse sequences imaged both in vitro and in vivo, but is sufficiently generic that it may be applied to nonneuronal cellular structures. We demonstrate the adaptability of GCA through the analysis of growth cone morphological variation and its relation to motility in both an unperturbed system and in the context of modified Rho GTPase signaling. We find that perturbations inducing similar changes in neurite length exhibit underappreciated phenotypic nuance at the scale of the growth cone.
Collapse
Affiliation(s)
- Maria M Bagonis
- Departments of Bioinformatics and Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX
- Department of Cell Biology, Harvard Medical School, Boston, MA
| | - Ludovico Fusco
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Olivier Pertz
- Department of Biomedicine, University of Basel, Basel, Switzerland
- Institute of Cell Biology, University of Bern, Bern, Switzerland
| | - Gaudenz Danuser
- Departments of Bioinformatics and Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX
- Department of Cell Biology, Harvard Medical School, Boston, MA
| |
Collapse
|
22
|
Lindblom RPF, Shen Q, Axén S, Landegren U, Kamali-Moghaddam M, Thelin S. Protein Profiling in Serum and Cerebrospinal Fluid Following Complex Surgery on the Thoracic Aorta Identifies Biological Markers of Neurologic Injury. J Cardiovasc Transl Res 2018; 11:503-516. [PMID: 30367354 PMCID: PMC6294830 DOI: 10.1007/s12265-018-9835-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 09/10/2018] [Indexed: 12/19/2022]
Abstract
Surgery on the arch or descending aorta is associated with significant risk of neurological complications. As a consequence of intubation and sedation, early neurologic injury may remain unnoticed. Biomarkers to aid in the initial diagnostics could prove of great value as immediate intervention is critical. Twenty-three patients operated in the thoracic aorta with significant risk of perioperative neurological injury were included. Cerebrospinal fluid (CSF) and serum were obtained preoperatively and in the first and second postoperative days and assessed with a panel of 92 neurological-related proteins. Three patients suffered spinal cord injury (SCI), eight delirium, and nine hallucinations. There were markers in both serum and CSF that differed between the affected and non-affected patients (SCI; IL6, GFAP, CSPG4, delirium; TR4, EZH2, hallucinations; NF1). The study identifies markers in serum and CSF that reflect the occurrence of neurologic insults following aortic surgery, which may aid in the care of these patients.
Collapse
Affiliation(s)
- Rickard P F Lindblom
- Department of Cardiothoracic Surgery and Anesthesia, Uppsala University Hospital, SE-751 85, Uppsala, Sweden. .,Department of Surgical Sciences, Section of Thoracic Surgery, Uppsala University, Uppsala, Sweden.
| | - Qiujin Shen
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Sofie Axén
- Department of Cardiothoracic Surgery and Anesthesia, Uppsala University Hospital, SE-751 85, Uppsala, Sweden
| | - Ulf Landegren
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Masood Kamali-Moghaddam
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Stefan Thelin
- Department of Cardiothoracic Surgery and Anesthesia, Uppsala University Hospital, SE-751 85, Uppsala, Sweden.,Department of Surgical Sciences, Section of Thoracic Surgery, Uppsala University, Uppsala, Sweden
| |
Collapse
|
23
|
Herrera E, Agudo-Barriuso M, Murcia-Belmonte V. Cranial Pair II: The Optic Nerves. Anat Rec (Hoboken) 2018; 302:428-445. [DOI: 10.1002/ar.23922] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 04/19/2017] [Accepted: 05/14/2017] [Indexed: 11/12/2022]
Affiliation(s)
- Eloísa Herrera
- Instituto de Neurociencias de Alicante (Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, CSIC-UMH); Av. Santiago Ramón y Cajal, s/n., 03550 Sant Joan d'Alacant Alicante Spain
| | - Marta Agudo-Barriuso
- Departamento de Oftalmología, Facultad de Medicina; Universidad de Murcia, Instituto Murciano de Investigación Biosanitaria-Virgen de la Arrixaca (IMIB-Arrixaca); Murcia Spain
| | - Verónica Murcia-Belmonte
- Instituto de Neurociencias de Alicante (Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, CSIC-UMH); Av. Santiago Ramón y Cajal, s/n., 03550 Sant Joan d'Alacant Alicante Spain
| |
Collapse
|
24
|
Fligor CM, Langer KB, Sridhar A, Ren Y, Shields PK, Edler MC, Ohlemacher SK, Sluch VM, Zack DJ, Zhang C, Suter DM, Meyer JS. Three-Dimensional Retinal Organoids Facilitate the Investigation of Retinal Ganglion Cell Development, Organization and Neurite Outgrowth from Human Pluripotent Stem Cells. Sci Rep 2018; 8:14520. [PMID: 30266927 PMCID: PMC6162218 DOI: 10.1038/s41598-018-32871-8] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 08/24/2018] [Indexed: 12/13/2022] Open
Abstract
Retinal organoids are three-dimensional structures derived from human pluripotent stem cells (hPSCs) which recapitulate the spatial and temporal differentiation of the retina, serving as effective in vitro models of retinal development. However, a lack of emphasis has been placed upon the development and organization of retinal ganglion cells (RGCs) within retinal organoids. Thus, initial efforts were made to characterize RGC differentiation throughout early stages of organoid development, with a clearly defined RGC layer developing in a temporally-appropriate manner expressing a complement of RGC-associated markers. Beyond studies of RGC development, retinal organoids may also prove useful for cellular replacement in which extensive axonal outgrowth is necessary to reach post-synaptic targets. Organoid-derived RGCs could help to elucidate factors promoting axonal outgrowth, thereby identifying approaches to circumvent a formidable obstacle to RGC replacement. As such, additional efforts demonstrated significant enhancement of neurite outgrowth through modulation of both substrate composition and growth factor signaling. Additionally, organoid-derived RGCs exhibited diverse phenotypes, extending elaborate growth cones and expressing numerous guidance receptors. Collectively, these results establish retinal organoids as a valuable tool for studies of RGC development, and demonstrate the utility of organoid-derived RGCs as an effective platform to study factors influencing neurite outgrowth from organoid-derived RGCs.
Collapse
Affiliation(s)
- Clarisse M Fligor
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis, IN, 46202, USA
| | - Kirstin B Langer
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis, IN, 46202, USA
| | - Akshayalakshmi Sridhar
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis, IN, 46202, USA
- Department of Biological Structure, University of Washington, Seattle, WA, 98195, USA
| | - Yuan Ren
- Department of Biological Sciences, Purdue University, West Lafayette, IN, 47907, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, 47907, USA
| | - Priya K Shields
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis, IN, 46202, USA
| | - Michael C Edler
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis, IN, 46202, USA
| | - Sarah K Ohlemacher
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis, IN, 46202, USA
| | - Valentin M Sluch
- Department of Molecular Biology and Genetics, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Donald J Zack
- Department of Molecular Biology and Genetics, Johns Hopkins University, Baltimore, MD, 21287, USA
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, 21287, USA
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, 21287, USA
- Institute of Genetic Medicine, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Chi Zhang
- Department of Medical and Molecular Genetics, Indiana University, Indianapolis, IN, 46202, USA
| | - Daniel M Suter
- Department of Biological Sciences, Purdue University, West Lafayette, IN, 47907, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, 47907, USA
| | - Jason S Meyer
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis, IN, 46202, USA.
- Department of Medical and Molecular Genetics, Indiana University, Indianapolis, IN, 46202, USA.
- Stark Neurosciences Research Institute, Indiana University, Indianapolis, IN, 46202, USA.
| |
Collapse
|
25
|
Cheng Z, Zou X, Jin Y, Gao S, Lv J, Li B, Cui R. The Role of KLF 4 in Alzheimer's Disease. Front Cell Neurosci 2018; 12:325. [PMID: 30297986 PMCID: PMC6160590 DOI: 10.3389/fncel.2018.00325] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Accepted: 09/07/2018] [Indexed: 01/30/2023] Open
Abstract
Krüppel-like factor 4 (KLF4), a member of the family of zinc-finger transcription factors, is widely expressed in range of tissues that play multiple functions. Emerging evidence suggest KLF4’s critical regulatory effect on the neurophysiological and neuropathological processes of Alzheimer’s disease (AD), indicating that KLF4 might be a potential therapeutic target of neurodegenerative diseases. In this review, we will summarize relevant studies and illuminate the regulatory role of KLF4 in the neuroinflammation, neuronal apoptosis, axon regeneration and iron accumulation to clarify KLF4’s status in the pathogenesis of AD.
Collapse
Affiliation(s)
- Ziqian Cheng
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Xiaohan Zou
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Yang Jin
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Shuohui Gao
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Jiayin Lv
- Department of Gastrointestinal Colorectal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Bingjin Li
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Ranji Cui
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
26
|
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: 256] [Impact Index Per Article: 42.7] [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
|
27
|
An Image-Based miRNA Screen Identifies miRNA-135s As Regulators of CNS Axon Growth and Regeneration by Targeting Krüppel-like Factor 4. J Neurosci 2017; 38:613-630. [PMID: 29196317 DOI: 10.1523/jneurosci.0662-17.2017] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 10/24/2017] [Accepted: 10/29/2017] [Indexed: 01/08/2023] Open
Abstract
During embryonic development, axons extend over long distances to establish functional connections. In contrast, axon regeneration in the adult mammalian CNS is limited in part by a reduced intrinsic capacity for axon growth. Therefore, insight into the intrinsic control of axon growth may provide new avenues for enhancing CNS regeneration. Here, we performed one of the first miRNome-wide functional miRNA screens to identify miRNAs with robust effects on axon growth. High-content screening identified miR-135a and miR-135b as potent stimulators of axon growth and cortical neuron migration in vitro and in vivo in male and female mice. Intriguingly, both of these developmental effects of miR-135s relied in part on silencing of Krüppel-like factor 4 (KLF4), a well known intrinsic inhibitor of axon growth and regeneration. These results prompted us to test the effect of miR-135s on axon regeneration after injury. Our results show that intravitreal application of miR-135s facilitates retinal ganglion cell (RGC) axon regeneration after optic nerve injury in adult mice in part by repressing KLF4. In contrast, depletion of miR-135s further reduced RGC axon regeneration. Together, these data identify a novel neuronal role for miR-135s and the miR-135-KLF4 pathway and highlight the potential of miRNAs as tools for enhancing CNS axon regeneration.SIGNIFICANCE STATEMENT Axon regeneration in the adult mammalian CNS is limited in part by a reduced intrinsic capacity for axon growth. Therefore, insight into the intrinsic control of axon growth may provide new avenues for enhancing regeneration. By performing an miRNome-wide functional screen, our studies identify miR-135s as stimulators of axon growth and neuron migration and show that intravitreal application of these miRNAs facilitates CNS axon regeneration after nerve injury in adult mice. Intriguingly, these developmental and regeneration-promoting effects rely in part on silencing of Krüppel-like factor 4 (KLF4), a well known intrinsic inhibitor of axon regeneration. Our data identify a novel neuronal role for the miR-135-KLF4 pathway and support the idea that miRNAs can be used for enhancing CNS axon regeneration.
Collapse
|
28
|
Bollaerts I, Veys L, Geeraerts E, Andries L, De Groef L, Buyens T, Salinas-Navarro M, Moons L, Van Hove I. Complementary research models and methods to study axonal regeneration in the vertebrate retinofugal system. Brain Struct Funct 2017; 223:545-567. [DOI: 10.1007/s00429-017-1571-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 11/15/2017] [Indexed: 01/18/2023]
|
29
|
Simmons AB, Bloomsburg SJ, Sukeena JM, Miller CJ, Ortega-Burgos Y, Borghuis BG, Fuerst PG. DSCAM-mediated control of dendritic and axonal arbor outgrowth enforces tiling and inhibits synaptic plasticity. Proc Natl Acad Sci U S A 2017; 114:E10224-E10233. [PMID: 29114051 PMCID: PMC5703318 DOI: 10.1073/pnas.1713548114] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Mature mammalian neurons have a limited ability to extend neurites and make new synaptic connections, but the mechanisms that inhibit such plasticity remain poorly understood. Here, we report that OFF-type retinal bipolar cells in mice are an exception to this rule, as they form new anatomical connections within their tiled dendritic fields well after retinal maturity. The Down syndrome cell-adhesion molecule (Dscam) confines these anatomical rearrangements within the normal tiled fields, as conditional deletion of the gene permits extension of dendrite and axon arbors beyond these borders. Dscam deletion in the mature retina results in expanded dendritic fields and increased cone photoreceptor contacts, demonstrating that DSCAM actively inhibits circuit-level plasticity. Electrophysiological recordings from Dscam-/- OFF bipolar cells showed enlarged visual receptive fields, demonstrating that expanded dendritic territories comprise functional synapses. Our results identify cell-adhesion molecule-mediated inhibition as a regulator of circuit-level neuronal plasticity in the adult retina.
Collapse
Affiliation(s)
- Aaron B Simmons
- Department of Biological Sciences, University of Idaho, Moscow, ID 83844
| | | | - Joshua M Sukeena
- Department of Biological Sciences, University of Idaho, Moscow, ID 83844
| | - Calvin J Miller
- Department of Biological Sciences, University of Idaho, Moscow, ID 83844
| | - Yohaniz Ortega-Burgos
- Department of Chemistry, University of Puerto Rico-Humacao, Humacao Puerto Rico, 00792
| | - Bart G Borghuis
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY 40202;
| | - Peter G Fuerst
- Department of Biological Sciences, University of Idaho, Moscow, ID 83844;
- Washington-Wyoming-Alaska-Montana-Idaho Medical Education Program, University of Washington School of Medicine, Moscow, ID 83844
| |
Collapse
|
30
|
Faust A, Kandakatla A, van der Merwe Y, Ren T, Huleihel L, Hussey G, Naranjo JD, Johnson S, Badylak S, Steketee M. Urinary bladder extracellular matrix hydrogels and matrix-bound vesicles differentially regulate central nervous system neuron viability and axon growth and branching. J Biomater Appl 2017; 31:1277-1295. [PMID: 28447547 DOI: 10.1177/0885328217698062] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Central nervous system neurons often degenerate after trauma due to the inflammatory innate immune response to injury, which can lead to neuronal cell death, scarring, and permanently lost neurologic function. Extracellular matrix bioscaffolds, derived by decellularizing healthy tissues, have been widely used in both preclinical and clinical studies to promote positive tissue remodeling, including neurogenesis, in numerous tissues, with extracellular matrix from homologous tissues often inducing more positive responses. Extracellular matrix hydrogels are liquid at room temperature and enable minimally invasive extracellular matrix injections into central nervous system tissues, before gelation at 37℃. However, few studies have analyzed how extracellular matrix hydrogels influence primary central nervous system neuron survival and growth, and whether central nervous system and non-central nervous system extracellular matrix specificity is critical to neuronal responses. Urinary bladder extracellular matrix hydrogels increase both primary hippocampal neuron survival and neurite growth to similar or even greater extents, suggesting extracellular matrix from non-homologous tissue sources, such as urinary bladder matrix-extracellular matrix, may be a more economical and safer alternative to developing central nervous system extracellular matrices for central nervous system applications. Additionally, we show matrix-bound vesicles derived from urinary bladder extracellular matrix are endocytosed by hippocampal neurons and positively regulate primary hippocampal neuron neurite growth. Matrix-bound vesicles carry protein and RNA cargos, including noncoding RNAs and miRNAs that map to the human genome and are known to regulate cellular processes. Thus, urinary bladder matrix-bound vesicles provide natural and transfectable cargoes which offer new experimental tools and therapeutic applications to study and treat central nervous system neuron injury.
Collapse
Affiliation(s)
- Anne Faust
- 1 Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,2 McGowan Institute for Regenerative Medicine, Pittsburgh, PA, USA
| | - Apoorva Kandakatla
- 1 Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,2 McGowan Institute for Regenerative Medicine, Pittsburgh, PA, USA
| | - Yolandi van der Merwe
- 1 Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,2 McGowan Institute for Regenerative Medicine, Pittsburgh, PA, USA.,3 Swanson School of Engineering, Department of Bioengineering University of Pittsburgh, Pittsburgh, PA, USA
| | - Tanchen Ren
- 1 Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,2 McGowan Institute for Regenerative Medicine, Pittsburgh, PA, USA
| | - Luai Huleihel
- 2 McGowan Institute for Regenerative Medicine, Pittsburgh, PA, USA.,4 Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - George Hussey
- 2 McGowan Institute for Regenerative Medicine, Pittsburgh, PA, USA.,4 Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Juan Diego Naranjo
- 2 McGowan Institute for Regenerative Medicine, Pittsburgh, PA, USA.,4 Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Scott Johnson
- 2 McGowan Institute for Regenerative Medicine, Pittsburgh, PA, USA.,4 Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Stephen Badylak
- 2 McGowan Institute for Regenerative Medicine, Pittsburgh, PA, USA.,4 Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Michael Steketee
- 1 Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,2 McGowan Institute for Regenerative Medicine, Pittsburgh, PA, USA.,5 Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
31
|
Feoktistov AI, Herman TG. Wallenda/DLK protein levels are temporally downregulated by Tramtrack69 to allow R7 growth cones to become stationary boutons. Development 2016; 143:2983-93. [PMID: 27402706 DOI: 10.1242/dev.134403] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 06/23/2016] [Indexed: 11/20/2022]
Abstract
Dual leucine zipper kinase (DLK) promotes growth cone motility and must be restrained to ensure normal development. PHR (Pam/Highwire/RPM-1) ubiquitin ligases therefore target DLK for degradation unless axon injury occurs. Overall DLK levels decrease during development, but how DLK levels are regulated within a developing growth cone has not been examined. We analyzed the expression of the fly DLK Wallenda (Wnd) in R7 photoreceptor growth cones as they halt at their targets and become presynaptic boutons. We found that Wnd protein levels are repressed by the PHR protein Highwire (Hiw) during R7 growth cone halting, as has been observed in other systems. However, as R7 growth cones become boutons, Wnd levels are further repressed by a temporally expressed transcription factor, Tramtrack69 (Ttk69). Previously unobserved negative feedback from JNK also contributes to Wnd repression at both time points. We conclude that neurons deploy additional mechanisms to downregulate DLK as they form stable, synaptic connections. We use live imaging to probe the effects of Wnd and Ttk69 on R7 bouton development and conclude that Ttk69 coordinates multiple regulators of this process.
Collapse
Affiliation(s)
- Alexander I Feoktistov
- Institute of Molecular Biology, Department of Biology, University of Oregon, Eugene, OR 97403, USA
| | - Tory G Herman
- Institute of Molecular Biology, Department of Biology, University of Oregon, Eugene, OR 97403, USA
| |
Collapse
|
32
|
Wang J, Galvao J, Beach KM, Luo W, Urrutia RA, Goldberg JL, Otteson DC. Novel Roles and Mechanism for Krüppel-like Factor 16 (KLF16) Regulation of Neurite Outgrowth and Ephrin Receptor A5 (EphA5) Expression in Retinal Ganglion Cells. J Biol Chem 2016; 291:18084-95. [PMID: 27402841 DOI: 10.1074/jbc.m116.732339] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Indexed: 11/06/2022] Open
Abstract
Regenerative medicine holds great promise for the treatment of degenerative retinal disorders. Krüppel-like factors (KLFs) are transcription factors that have recently emerged as key tools in regenerative medicine because some of them can function as epigenetic reprogrammers in stem cell biology. Here, we show that KLF16, one of the least understood members of this family, is a POU4F2 independent transcription factor in retinal ganglion cells (RGCs) as early as embryonic day 15. When overexpressed, KLF16 inhibits RGC neurite outgrowth and enhances RGC growth cone collapse in response to exogenous ephrinA5 ligands. Ephrin/EPH signaling regulates RGC connectivity. The EphA5 promoter contains multiple GC- and GT-rich KLF-binding sites, which, as shown by ChIP-assays, bind KLF16 in vivo In electrophoretic mobility shift assays, KLF16 binds specifically to a single KLF site near the EphA5 transcription start site that is required for KLF16 transactivation. Interestingly, methylation of only six of 98 CpG dinucleotides within the EphA5 promoter blocks its transactivation by KLF16 but enables transactivation by KLF2 and KLF15. These data demonstrate a role for KLF16 in regulation of RGC neurite outgrowth and as a methylation-sensitive transcriptional regulator of EphA5 expression. Together, these data identify differential low level methylation as a novel mechanism for regulating KLF16-mediated EphA5 expression across the retina. Because of the critical role of ephrin/EPH signaling in patterning RGC connectivity, understanding the role of KLFs in regulating neurite outgrowth and Eph receptor expression will be vital for successful restoration of functional vision through optic nerve regenerative therapies.
Collapse
Affiliation(s)
- Jianbo Wang
- From the Departments of Physiological Optics and Vision Science and
| | - Joana Galvao
- the Byers Eye Institute, School of Medicine, Stanford University, Palo Alto, California 94303, the Shiley Eye Institute, University of California San Diego, La Jolla, California 92093, and
| | - Krista M Beach
- From the Departments of Physiological Optics and Vision Science and
| | - Weijia Luo
- Biology and Biochemistry, University of Houston, Houston, Texas 77204
| | - Raul A Urrutia
- the Laboratory of Epigenetics and Chromatin Dynamics, Gastroenterology Research Unit, Epigenomics Translational Program, Center for Individualized Medicine, Departments of Medicine, Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota 55905
| | - Jeffrey L Goldberg
- the Byers Eye Institute, School of Medicine, Stanford University, Palo Alto, California 94303, the Shiley Eye Institute, University of California San Diego, La Jolla, California 92093, and
| | - Deborah C Otteson
- From the Departments of Physiological Optics and Vision Science and Biology and Biochemistry, University of Houston, Houston, Texas 77204,
| |
Collapse
|
33
|
Bhattarai S, Sochacka-Marlowe A, Crutchfield G, Khan R, Londraville R, Liu Q. Krüpple-like factors 7 and 6a mRNA expression in adult zebrafish central nervous system. Gene Expr Patterns 2016; 21:41-53. [PMID: 27364471 DOI: 10.1016/j.gep.2016.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 06/15/2016] [Accepted: 06/18/2016] [Indexed: 11/25/2022]
Abstract
Krüpple-like factors (KLFs) are transcription factors with zinc finger DNA binding domains known to play important roles in brain development and central nervous system (CNS) regeneration. There is little information on KLFs expression in adult vertebrate CNS. In this study, we used in situ hybridization to examine Klf7 mRNA (klf7) and Klf6a mRNA (klf6a) expression in adult zebrafish CNS. Both klfs exhibit wide and similar expression in the zebrafish CNS. Brain areas containing strongly labeled cells include the ventricular regions of the dorsomedial telencephalon, the ventromedial telencephalon, periventricular regions of the thalamus and hypothalamus, torus longitudinalis, stratum periventriculare of the optic tectum, granular regions of the cerebellar body and valvula, and superficial layers of the facial and vagal lobes. In the spinal cord, klf7- and klf6a-expressing cells are found in both the dorsal and ventral horns. Numerous sensory structures (e.g. auditory, lateral line, olfactory and visual) and several motor nuclei (e.g. oculomotor, trigeminal, and vagal motor nuclei) contain klf7- and/or klf6a-expressing cells. Our results may provide useful information for determining these Klfs in maintenance and/or function in adult CNS.
Collapse
Affiliation(s)
- Sunil Bhattarai
- Department of Biology and Integrated Bioscience Program, University of Akron, Akron, OH 44325, United States
| | - Alicja Sochacka-Marlowe
- Department of Biology and Integrated Bioscience Program, University of Akron, Akron, OH 44325, United States
| | - Gerald Crutchfield
- Department of Biology and Integrated Bioscience Program, University of Akron, Akron, OH 44325, United States
| | - Ramisha Khan
- Department of Biology and Integrated Bioscience Program, University of Akron, Akron, OH 44325, United States
| | - Richard Londraville
- Department of Biology and Integrated Bioscience Program, University of Akron, Akron, OH 44325, United States
| | - Qin Liu
- Department of Biology and Integrated Bioscience Program, University of Akron, Akron, OH 44325, United States.
| |
Collapse
|
34
|
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: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
35
|
van Niekerk EA, Tuszynski MH, Lu P, Dulin JN. Molecular and Cellular Mechanisms of Axonal Regeneration After Spinal Cord Injury. Mol Cell Proteomics 2015; 15:394-408. [PMID: 26695766 DOI: 10.1074/mcp.r115.053751] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Indexed: 12/28/2022] Open
Abstract
Following axotomy, a complex temporal and spatial coordination of molecular events enables regeneration of the peripheral nerve. In contrast, multiple intrinsic and extrinsic factors contribute to the general failure of axonal regeneration in the central nervous system. In this review, we examine the current understanding of differences in protein expression and post-translational modifications, activation of signaling networks, and environmental cues that may underlie the divergent regenerative capacity of central and peripheral axons. We also highlight key experimental strategies to enhance axonal regeneration via modulation of intraneuronal signaling networks and the extracellular milieu. Finally, we explore potential applications of proteomics to fill gaps in the current understanding of molecular mechanisms underlying regeneration, and to provide insight into the development of more effective approaches to promote axonal regeneration following injury to the nervous system.
Collapse
Affiliation(s)
- Erna A van Niekerk
- From the ‡Department of Neurosciences, University of California, San Diego, La Jolla, CA, 92093;
| | - Mark H Tuszynski
- From the ‡Department of Neurosciences, University of California, San Diego, La Jolla, CA, 92093; §Veterans Administration Medical Center, San Diego, CA 92161
| | - Paul Lu
- From the ‡Department of Neurosciences, University of California, San Diego, La Jolla, CA, 92093; §Veterans Administration Medical Center, San Diego, CA 92161
| | - Jennifer N Dulin
- From the ‡Department of Neurosciences, University of California, San Diego, La Jolla, CA, 92093
| |
Collapse
|
36
|
Yu J, Luan X, Lan S, Yan B, Maier A. Fasudil, a Rho-Associated Protein Kinase Inhibitor, Attenuates Traumatic Retinal Nerve Injury in Rabbits. J Mol Neurosci 2015; 58:74-82. [DOI: 10.1007/s12031-015-0691-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Accepted: 11/23/2015] [Indexed: 12/17/2022]
|
37
|
A novel perspective on neuron study: damaging and promoting effects in different neurons induced by mechanical stress. Biomech Model Mechanobiol 2015; 15:1019-27. [DOI: 10.1007/s10237-015-0743-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 10/29/2015] [Indexed: 12/11/2022]
|
38
|
Heine P, Ehrlicher A, Käs J. Neuronal and metastatic cancer cells: Unlike brothers. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:3126-31. [DOI: 10.1016/j.bbamcr.2015.06.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 06/10/2015] [Accepted: 06/12/2015] [Indexed: 12/22/2022]
|
39
|
Heidemann SR, Bray D. Tension-driven axon assembly: a possible mechanism. Front Cell Neurosci 2015; 9:316. [PMID: 26321917 PMCID: PMC4532915 DOI: 10.3389/fncel.2015.00316] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 07/29/2015] [Indexed: 12/03/2022] Open
Affiliation(s)
- Steven R Heidemann
- Department of Physiology, Michigan State University East Lansing, MI, USA
| | - Dennis Bray
- Department of Physiology, Development and Neuroscience, University of Cambridge Cambridge, UK
| |
Collapse
|
40
|
Pita-Thomas W, Steketee MB, Moysidis SN, Thakor K, Hampton B, Goldberg JL. Promoting filopodial elongation in neurons by membrane-bound magnetic nanoparticles. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2015; 11:559-67. [PMID: 25596077 DOI: 10.1016/j.nano.2014.11.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Revised: 10/30/2014] [Accepted: 11/24/2014] [Indexed: 12/15/2022]
Abstract
Filopodia are 5-10 μm long processes that elongate by actin polymerization, and promote axon growth and guidance by exerting mechanical tension and by molecular signaling. Although axons elongate in response to mechanical tension, the structural and functional effects of tension specifically applied to growth cone filopodia are unknown. Here we developed a strategy to apply tension specifically to retinal ganglion cell (RGC) growth cone filopodia through surface-functionalized, membrane-targeted superparamagnetic iron oxide nanoparticles (SPIONs). When magnetic fields were applied to surface-bound SPIONs, RGC filopodia elongated directionally, contained polymerized actin filaments, and generated retrograde forces, behaving as bona fide filopodia. Data presented here support the premise that mechanical tension induces filopodia growth but counter the hypothesis that filopodial tension directly promotes growth cone advance. Future applications of these approaches may be used to induce sustained forces on multiple filopodia or other subcellular microstructures to study axon growth or cell migration. From the clinical editor: Mechanical tension to the tip of filopodia is known to promote axonal growth. In this article, the authors used superparamagnetic iron oxide nanoparticles (SPIONs) targeted specifically to membrane molecules, then applied external magnetic field to elicit filopodial elongation, which provided a tool to study the role of mechanical forces in filopodia dynamics and function.
Collapse
Affiliation(s)
- Wolfgang Pita-Thomas
- Bascom Palmer Eye Institute and Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA; Department of Anatomy and Neurobiology, Washington University, St. Louis, MO, USA
| | - Michael B Steketee
- Bascom Palmer Eye Institute and Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA; Department of Ophthalmology and McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Stavros N Moysidis
- Bascom Palmer Eye Institute and Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Kinjal Thakor
- Bascom Palmer Eye Institute and Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Blake Hampton
- Bascom Palmer Eye Institute and Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jeffrey L Goldberg
- Bascom Palmer Eye Institute and Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA; Department of Ophthalmology, Shiley Eye Center, UC San Diego, San Diego, CA, USA.
| |
Collapse
|