51
|
Abstract
The field of microfluidics allows for the precise spatial manipulation of small amounts of fluids. Within microstructures, laminar flow of fluids can be exploited to control the diffusion of small molecules, creating desired microenvironments for cells. Cellular neuroscience has benefited greatly from devices designed to fluidically isolate cell bodies and axons. Microfluidic devices specialized for neuron compartmentalization are made of polydimethylsiloxane (PDMS) which is gas permeable, is compatible with fluorescence microscopy, and has low cost. These devices are commonly used to study signals initiated exclusively on axons, somatodendritic compartments, or even single synapses. We have also found that microfluidic devices allow for rapid, reproducible interrogation of axon degeneration. Here, we describe the methodology for assessing axonal degeneration in microfluidic devices. We describe several use cases, including enucleation (removal of cell bodies) and trophic deprivation to investigate axon degeneration in pathological and developmental scenarios, respectively.
Collapse
Affiliation(s)
- Yu Yong
- Department of Biology, University of Virginia, Charlottesville, VA, USA
| | - Christopher Hughes
- Department of Physics and Astronomy, James Madison University, Harrisonburg, VA, USA
| | | |
Collapse
|
52
|
Choi JH, Santhosh M, Choi JW. In Vitro Blood-Brain Barrier-Integrated Neurological Disorder Models Using a Microfluidic Device. MICROMACHINES 2019; 11:E21. [PMID: 31878184 PMCID: PMC7019695 DOI: 10.3390/mi11010021] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 12/20/2019] [Accepted: 12/21/2019] [Indexed: 12/14/2022]
Abstract
The blood-brain barrier (BBB) plays critical role in the human physiological system such as protection of the central nervous system (CNS) from external materials in the blood vessel, including toxicants and drugs for several neurological disorders, a critical type of human disease. Therefore, suitable in vitro BBB models with fluidic flow to mimic the shear stress and supply of nutrients have been developed. Neurological disorder has also been investigated for developing realistic models that allow advance fundamental and translational research and effective therapeutic strategy design. Here, we discuss introduction of the blood-brain barrier in neurological disorder models by leveraging a recently developed microfluidic system and human organ-on-a-chip system. Such models could provide an effective drug screening platform and facilitate personalized therapy of several neurological diseases.
Collapse
Affiliation(s)
- Jin-Ha Choi
- Department of Chemical and Biomolecular Engineering, Sogang University, 35 Baekbeom-ro (Sinsu-dong), Mapo-gu, 121-742 Seoul, Korea;
| | - Mallesh Santhosh
- Center for Integrated Biotechnology, Sogang University, 35 Baekbeom-ro (Sinsu-dong), Mapo-gu, 121-742 Seoul, Korea;
| | - Jeong-Woo Choi
- Department of Chemical and Biomolecular Engineering, Sogang University, 35 Baekbeom-ro (Sinsu-dong), Mapo-gu, 121-742 Seoul, Korea;
- Center for Integrated Biotechnology, Sogang University, 35 Baekbeom-ro (Sinsu-dong), Mapo-gu, 121-742 Seoul, Korea;
| |
Collapse
|
53
|
Hao Y, Waller TJ, Nye DM, Li J, Zhang Y, Hume RI, Rolls MM, Collins CA. Degeneration of Injured Axons and Dendrites Requires Restraint of a Protective JNK Signaling Pathway by the Transmembrane Protein Raw. J Neurosci 2019; 39:8457-8470. [PMID: 31492772 PMCID: PMC6807270 DOI: 10.1523/jneurosci.0016-19.2019] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 08/08/2019] [Accepted: 08/22/2019] [Indexed: 12/15/2022] Open
Abstract
The degeneration of injured axons involves a self-destruction pathway whose components and mechanism are not fully understood. Here, we report a new regulator of axonal resilience. The transmembrane protein Raw is cell autonomously required for the degeneration of injured axons, dendrites, and synapses in Drosophila melanogaster In both male and female raw hypomorphic mutant or knock-down larvae, the degeneration of injured axons, dendrites, and synapses from motoneurons and sensory neurons is strongly inhibited. This protection is insensitive to reduction in the levels of the NAD+ synthesis enzyme Nmnat (nicotinamide mononucleotide adenylyl transferase), but requires the c-Jun N-terminal kinase (JNK) mitogen-activated protein (MAP) kinase and the transcription factors Fos and Jun (AP-1). Although these factors were previously known to function in axonal injury signaling and regeneration, Raw's function can be genetically separated from other axonal injury responses: Raw does not modulate JNK-dependent axonal injury signaling and regenerative responses, but instead restrains a protective pathway that inhibits the degeneration of axons, dendrites, and synapses. Although protection in raw mutants requires JNK, Fos, and Jun, JNK also promotes axonal degeneration. These findings suggest the existence of multiple independent pathways that share modulation by JNK, Fos, and Jun that influence how axons respond to stress and injury.SIGNIFICANCE STATEMENT Axonal degeneration is a major feature of neuropathies and nerve injuries and occurs via a cell autonomous self-destruction pathway whose mechanism is poorly understood. This study reports the identification of a new regulator of axonal degeneration: the transmembrane protein Raw. Raw regulates a cell autonomous nuclear signaling pathway whose yet unknown downstream effectors protect injured axons, dendrites, and synapses from degenerating. These findings imply that the susceptibility of axons to degeneration is strongly regulated in neurons. Future understanding of the cellular pathway regulated by Raw, which engages the c-Jun N-terminal kinase (JNK) mitogen-activated protein (MAP) kinase and Fos and Jun transcription factors, may suggest new strategies to increase the resiliency of axons in debilitating neuropathies.
Collapse
Affiliation(s)
- Yan Hao
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan 48109-1085
| | - Thomas J Waller
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan 48109-1085
| | - Derek M Nye
- Huck Institutes of the Life Sciences, and Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania 16802, and
| | - Jiaxing Li
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan 48109-1085
| | - Yanxiao Zhang
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan 48109-2218
| | - Richard I Hume
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan 48109-1085
| | - Melissa M Rolls
- Huck Institutes of the Life Sciences, and Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania 16802, and
| | - Catherine A Collins
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan 48109-1085,
| |
Collapse
|
54
|
Llobet Rosell A, Neukomm LJ. Axon death signalling in Wallerian degeneration among species and in disease. Open Biol 2019; 9:190118. [PMID: 31455157 PMCID: PMC6731592 DOI: 10.1098/rsob.190118] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Axon loss is a shared feature of nervous systems being challenged in neurological disease, by chemotherapy or mechanical force. Axons take up the vast majority of the neuronal volume, thus numerous axonal intrinsic and glial extrinsic support mechanisms have evolved to promote lifelong axonal survival. Impaired support leads to axon degeneration, yet underlying intrinsic signalling cascades actively promoting the disassembly of axons remain poorly understood in any context, making the development to attenuate axon degeneration challenging. Wallerian degeneration serves as a simple model to study how axons undergo injury-induced axon degeneration (axon death). Severed axons actively execute their own destruction through an evolutionarily conserved axon death signalling cascade. This pathway is also activated in the absence of injury in diseased and challenged nervous systems. Gaining insights into mechanisms underlying axon death signalling could therefore help to define targets to block axon loss. Herein, we summarize features of axon death at the molecular and subcellular level. Recently identified and characterized mediators of axon death signalling are comprehensively discussed in detail, and commonalities and differences across species highlighted. We conclude with a summary of engaged axon death signalling in humans and animal models of neurological conditions. Thus, gaining mechanistic insights into axon death signalling broadens our understanding beyond a simple injury model. It harbours the potential to define targets for therapeutic intervention in a broad range of human axonopathies.
Collapse
Affiliation(s)
- Arnau Llobet Rosell
- Department of Fundamental Neurosciences, University of Lausanne, Rue du Bugnon 9, 1005 Lausanne, VD, Switzerland
| | - Lukas J Neukomm
- Department of Fundamental Neurosciences, University of Lausanne, Rue du Bugnon 9, 1005 Lausanne, VD, Switzerland
| |
Collapse
|
55
|
Keilhoff G, Nguyen Thi TM, Esser T, Ebmeyer U. Relative Resilience of Cerebellar Purkinje Cells in a Cardiac Arrest/Resuscitation Rat Model. Neurocrit Care 2019; 32:775-789. [DOI: 10.1007/s12028-019-00799-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
56
|
Datar A, Ameeramja J, Bhat A, Srivastava R, Mishra A, Bernal R, Prost J, Callan-Jones A, Pullarkat PA. The Roles of Microtubules and Membrane Tension in Axonal Beading, Retraction, and Atrophy. Biophys J 2019; 117:880-891. [PMID: 31427070 DOI: 10.1016/j.bpj.2019.07.046] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 06/23/2019] [Accepted: 07/26/2019] [Indexed: 02/02/2023] Open
Abstract
Axonal beading, or the formation of a series of swellings along the axon, and retraction are commonly observed shape transformations that precede axonal atrophy in Alzheimer's disease, Parkinson's disease, and other neurodegenerative conditions. The mechanisms driving these morphological transformations are poorly understood. Here, we report controlled experiments that can induce either beading or retraction and follow the time evolution of these responses. By making quantitative analysis of the shape modes under different conditions, measurement of membrane tension, and using theoretical considerations, we argue that membrane tension is the main driving force that pushes cytosol out of the axon when microtubules are degraded, causing axonal thinning. Under pharmacological perturbation, atrophy is always retrograde, and this is set by a gradient in the microtubule stability. The nature of microtubule depolymerization dictates the type of shape transformation, vis-à-vis beading or retraction. Elucidating the mechanisms of these shape transformations may facilitate development of strategies to prevent or arrest axonal atrophy due to neurodegenerative conditions.
Collapse
Affiliation(s)
| | | | - Alka Bhat
- Raman Research Institute, Bengaluru, India
| | | | | | - Roberto Bernal
- Departamento de Física, SMAT-C, Universidad de Santiago de Chile, Santiago, Chile
| | - Jacques Prost
- Laboratoire Physico Chimie Curie, Institut Curie, 10 PSL Research University, CNRS UMR168, Paris, France; Mechanobiology Institute, National University of Singapore, Singapore
| | - Andrew Callan-Jones
- Laboratoire Matière et Systèmes Complexes, Université Paris Diderot, Paris, France.
| | | |
Collapse
|
57
|
Shao X, You R, Hui TH, Fang C, Gong Z, Yan Z, Chang RCC, Shenoy VB, Lin Y. Tension- and Adhesion-Regulated Retraction of Injured Axons. Biophys J 2019; 117:193-202. [PMID: 31278003 DOI: 10.1016/j.bpj.2019.06.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 05/27/2019] [Accepted: 06/14/2019] [Indexed: 12/16/2022] Open
Abstract
Damage-induced retraction of axons during traumatic brain injury is believed to play a key role in the disintegration of the neural network and to eventually lead to severe symptoms such as permanent memory loss and emotional disturbances. However, fundamental questions such as how axon retraction progresses and what physical factors govern this process still remain unclear. Here, we report a combined experimental and modeling study to address these questions. Specifically, a sharp atomic force microscope probe was used to transect axons and trigger their retraction in a precisely controlled manner. Interestingly, we showed that the retracting motion of a well-developed axon can be arrested by strong cell-substrate attachment. However, axon retraction was found to be retriggered if a second transection was conducted, albeit with a lower shrinking amplitude. Furthermore, disruption of the actin cytoskeleton or cell-substrate adhesion significantly altered the retracting dynamics of injured axons. Finally, a mathematical model was developed to explain the observed injury response of neural cells in which the retracting motion was assumed to be driven by the pre-tension in the axon and progress against neuron-substrate adhesion as well as the viscous resistance of the cell. Using realistic parameters, model predictions were found to be in good agreement with our observations under a variety of experimental conditions. By revealing the essential physics behind traumatic axon retraction, findings here could provide insights on the development of treatment strategies for axonal injury as well as its possible interplay with other neurodegenerative diseases.
Collapse
Affiliation(s)
- Xueying Shao
- Department of Mechanical Engineering, The University of Hong Kong, Hong Kong, China; HKU-Shenzhen Institute of Research and Innovation, Shenzhen, Guangdong, China
| | - Ran You
- Laboratory of Neurodegenerative Diseases, School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Tsz Hin Hui
- Department of Mechanical Engineering, The University of Hong Kong, Hong Kong, China; HKU-Shenzhen Institute of Research and Innovation, Shenzhen, Guangdong, China
| | - Chao Fang
- Department of Mechanical Engineering, The University of Hong Kong, Hong Kong, China; HKU-Shenzhen Institute of Research and Innovation, Shenzhen, Guangdong, China
| | - Ze Gong
- Center for Engineering Mechanobiology and Department of Materials Science and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Zishen Yan
- Department of Mechanical Engineering, The University of Hong Kong, Hong Kong, China; HKU-Shenzhen Institute of Research and Innovation, Shenzhen, Guangdong, China
| | - Raymond Chuen Chung Chang
- Laboratory of Neurodegenerative Diseases, School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Vivek B Shenoy
- Center for Engineering Mechanobiology and Department of Materials Science and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania.
| | - Yuan Lin
- Department of Mechanical Engineering, The University of Hong Kong, Hong Kong, China; HKU-Shenzhen Institute of Research and Innovation, Shenzhen, Guangdong, China.
| |
Collapse
|
58
|
Pieper AA, McKnight SL. Benefits of Enhancing Nicotinamide Adenine Dinucleotide Levels in Damaged or Diseased Nerve Cells. COLD SPRING HARBOR SYMPOSIA ON QUANTITATIVE BIOLOGY 2019; 83:207-217. [PMID: 30787047 DOI: 10.1101/sqb.2018.83.037622] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Three unbiased lines of research have commonly pointed to the benefits of enhanced levels of nicotinamide adenine dinucleotide (NAD+) to diseased or damaged neurons. Mice carrying a triplication of the gene encoding the culminating enzyme in NAD+ salvage from nicotinamide, NMNAT, are protected from a variety of insults to axons. Protection from Wallerian degeneration of axons is also observed in flies and mice bearing inactivating mutations in the SARM1 gene. Functional studies of the SARM1 gene product have revealed the presence of an enzymatic activity directed toward the hydrolysis of NAD+ Finally, an unbiased drug screen performed in living mice led to the discovery of a neuroprotective chemical designated P7C3. Biochemical studies of the P7C3 chemical show that it can enhance recovery of NAD+ from nicotinamide by activating NAMPT, the first enzyme in the salvage pathway. In combination, these three unrelated research endeavors offer evidence of the benefits of enhanced NAD+ levels to damaged neurons.
Collapse
Affiliation(s)
- Andrew A Pieper
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, Ohio 44106, USA
- Department of Psychiatry, Case Western Reserve University, Cleveland, Ohio 44106, USA
- Geriatric Research Education and Clinical Centers, Louis Stokes Cleveland VAMC, Cleveland, Ohio 44106, USA
| | - Steven L McKnight
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| |
Collapse
|
59
|
Wang H, Wang X, Zhang K, Wang Q, Cao X, Wang Z, Zhang S, Li A, Liu K, Fang Y. Rapid depletion of ESCRT protein Vps4 underlies injury-induced autophagic impediment and Wallerian degeneration. SCIENCE ADVANCES 2019; 5:eaav4971. [PMID: 30788439 PMCID: PMC6374107 DOI: 10.1126/sciadv.aav4971] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 01/02/2019] [Indexed: 06/01/2023]
Abstract
Injured axons undergo a controlled, self-destruction process, known as Wallerian degeneration. However, the underlying mechanism remains elusive. Using the Drosophila wing nerve as a model, we identify the ESCRT component Vps4 as a previously unidentified essential gene for axonal integrity. Up-regulation of Vps4 remarkably delays degeneration of injured axons. We further reveal that Vps4 is required and sufficient to promote autophagic flux in axons and mammalian cells. Moreover, using both in vitro and in vivo models, we show that the function of Vps4 in maintaining axonal autophagy and suppressing Wallerian degeneration is conserved in mammals. Last, we uncover that Vps4 protein is rapidly depleted in injured mouse axons, which may underlie the injury-induced autophagic impediment and the subsequent axonal degeneration. Together, Vps4 and ESCRT may represent a novel signal transduction mechanism in axon injury and Wallerian degeneration.
Collapse
Affiliation(s)
- Haiqiong Wang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xuejie Wang
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
- Center of Systems Biology and Human Health, School of Science and Institute for Advanced Study, The Hong Kong University of Science and Technology, Hong Kong, China
- Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University–The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, China
| | - Kai Zhang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qingyao Wang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xu Cao
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhao Wang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
| | - Shuang Zhang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
| | - Ang Li
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Joint International Research Laboratory of CNS Regeneration Ministry of Education, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Jinan University, Guangzhou 510632, China
| | - Kai Liu
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
- Center of Systems Biology and Human Health, School of Science and Institute for Advanced Study, The Hong Kong University of Science and Technology, Hong Kong, China
- Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University–The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, China
| | - Yanshan Fang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
60
|
Davies AJ, Kim HW, Gonzalez-Cano R, Choi J, Back SK, Roh SE, Johnson E, Gabriac M, Kim MS, Lee J, Lee JE, Kim YS, Bae YC, Kim SJ, Lee KM, Na HS, Riva P, Latremoliere A, Rinaldi S, Ugolini S, Costigan M, Oh SB. Natural Killer Cells Degenerate Intact Sensory Afferents following Nerve Injury. Cell 2019; 176:716-728.e18. [PMID: 30712871 PMCID: PMC6418410 DOI: 10.1016/j.cell.2018.12.022] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 11/12/2018] [Accepted: 12/14/2018] [Indexed: 02/06/2023]
Abstract
Sensory axons degenerate following separation from their cell body, but partial injury to peripheral nerves may leave the integrity of damaged axons preserved. We show that an endogenous ligand for the natural killer (NK) cell receptor NKG2D, Retinoic Acid Early 1 (RAE1), is re-expressed in adult dorsal root ganglion neurons following peripheral nerve injury, triggering selective degeneration of injured axons. Infiltration of cytotoxic NK cells into the sciatic nerve by extravasation occurs within 3 days following crush injury. Using a combination of genetic cell ablation and cytokine-antibody complex stimulation, we show that NK cell function correlates with loss of sensation due to degeneration of injured afferents and reduced incidence of post-injury hypersensitivity. This neuro-immune mechanism of selective NK cell-mediated degeneration of damaged but intact sensory axons complements Wallerian degeneration and suggests the therapeutic potential of modulating NK cell function to resolve painful neuropathy through the clearance of partially damaged nerves. Cytotoxic NK cells infiltrate the damaged peripheral nerve within days of injury Injured sensory axons express NKG2D ligand RAE1 to signal degeneration by NK cells Clearance of damaged axons reduces development of chronic pain after nerve injury NK cells complement Wallerian degeneration to aid functional regeneration of PNS
Collapse
Affiliation(s)
- Alexander J Davies
- Dental Research Institute and Department of Neurobiology and Physiology, School of Dentistry, Seoul National University, Seoul 03080, Republic of Korea; Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Hyoung Woo Kim
- Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Rafael Gonzalez-Cano
- Departments of Anesthesia and Neurobiology, Children's Hospital Boston and Harvard Medical School, Boston, MA 02115, USA
| | - Jahyang Choi
- Dental Research Institute and Department of Neurobiology and Physiology, School of Dentistry, Seoul National University, Seoul 03080, Republic of Korea
| | - Seung Keun Back
- Departments of Physiology, Biochemistry and Molecular Biology, College of Medicine, Korea University, Seoul 02841, Republic of Korea
| | - Seung Eon Roh
- Department of Physiology, Seoul National University College of Medicine, Seoul 03087, Republic of Korea
| | - Errin Johnson
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK
| | - Melanie Gabriac
- Aix Marseille Univ, CNRS, INSERM, CIML, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Mi-Sun Kim
- Dental Research Institute and Department of Neurobiology and Physiology, School of Dentistry, Seoul National University, Seoul 03080, Republic of Korea
| | - Jaehee Lee
- Dental Research Institute and Department of Neurobiology and Physiology, School of Dentistry, Seoul National University, Seoul 03080, Republic of Korea; Departments of Physiology, Biochemistry and Molecular Biology, College of Medicine, Korea University, Seoul 02841, Republic of Korea
| | - Jeong Eun Lee
- Departments of Physiology, Biochemistry and Molecular Biology, College of Medicine, Korea University, Seoul 02841, Republic of Korea
| | - Yun Sook Kim
- Department of Anatomy and Neurobiology, School of Dentistry, Kyungpook National University, Daegu 700-412, Korea
| | - Yong Chul Bae
- Department of Anatomy and Neurobiology, School of Dentistry, Kyungpook National University, Daegu 700-412, Korea
| | - Sang Jeong Kim
- Department of Physiology, Seoul National University College of Medicine, Seoul 03087, Republic of Korea
| | - Kyung-Mi Lee
- Departments of Physiology, Biochemistry and Molecular Biology, College of Medicine, Korea University, Seoul 02841, Republic of Korea
| | - Heung Sik Na
- Departments of Physiology, Biochemistry and Molecular Biology, College of Medicine, Korea University, Seoul 02841, Republic of Korea
| | - Priscilla Riva
- Departments of Anesthesia and Neurobiology, Children's Hospital Boston and Harvard Medical School, Boston, MA 02115, USA
| | - Alban Latremoliere
- Neurosurgery Department, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Simon Rinaldi
- Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Sophie Ugolini
- Aix Marseille Univ, CNRS, INSERM, CIML, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Michael Costigan
- Departments of Anesthesia and Neurobiology, Children's Hospital Boston and Harvard Medical School, Boston, MA 02115, USA.
| | - Seog Bae Oh
- Dental Research Institute and Department of Neurobiology and Physiology, School of Dentistry, Seoul National University, Seoul 03080, Republic of Korea; Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
61
|
Umeda K, Negishi M, Katoh H. RasGRF1 mediates brain-derived neurotrophic factor-induced axonal growth in primary cultured cortical neurons. Biochem Biophys Rep 2018; 17:56-64. [PMID: 30582008 PMCID: PMC6295856 DOI: 10.1016/j.bbrep.2018.11.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 11/29/2018] [Indexed: 12/01/2022] Open
Abstract
The appropriate development and regulation of neuronal morphology are important to establish functional neuronal circuits and enable higher brain function of the central nervous system. R-Ras, a member of the Ras family of small GTPases, plays crucial roles in the regulation of axonal morphology, including outgrowth, branching, and guidance. GTP-bound activated R-Ras reorganizes actin filaments and microtubules through interactions with its downstream effectors, leading to the precise control of axonal morphology. However, little is known about the upstream regulatory mechanisms for R-Ras activation in neurons. In this study, we found that brain-derived neurotrophic factor (BDNF) has a positive effect on endogenous R-Ras activation and promotes R-Ras-mediated axonal growth. RNA interference knockdown and overexpression experiments revealed that RasGRF1, a guanine nucleotide exchange factor (GEF) for R-Ras, is involved in BDNF-induced R-Ras activation and the promotion of axonal growth. Phosphorylation of RasGRF1 by protein kinase A at Ser916/898 is needed for the full activation of its GEF activity and to facilitate Ras signaling. We observed that BDNF treatment markedly increased this phosphorylation. Our results suggest that BDNF is one of the critical extrinsic regulators for R-Ras activation, and that RasGRF1 is an intrinsic key mediator for BDNF-induced R-Ras activation and R-Ras-mediated axonal morphological regulation.
Collapse
Affiliation(s)
- Kentaro Umeda
- Laboratory of Molecular Neurobiology, Graduate School of Pharmaceutical Sciences, Kyoto University, Yoshidakonoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Manabu Negishi
- Laboratory of Molecular Neurobiology, Graduate School of Pharmaceutical Sciences, Kyoto University, Yoshidakonoe-cho, Sakyo-ku, Kyoto 606-8501, Japan.,Laboratory of Molecular Neurobiology, Graduate School of Biostudies, Kyoto University, Yoshidakonoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Hironori Katoh
- Laboratory of Molecular Neurobiology, Graduate School of Pharmaceutical Sciences, Kyoto University, Yoshidakonoe-cho, Sakyo-ku, Kyoto 606-8501, Japan.,Laboratory of Molecular Neurobiology, Graduate School of Biostudies, Kyoto University, Yoshidakonoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| |
Collapse
|
62
|
Neumann B, Linton C, Giordano-Santini R, Hilliard MA. Axonal fusion: An alternative and efficient mechanism of nerve repair. Prog Neurobiol 2018; 173:88-101. [PMID: 30500382 DOI: 10.1016/j.pneurobio.2018.11.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 11/22/2018] [Accepted: 11/26/2018] [Indexed: 02/07/2023]
Abstract
Injuries to the nervous system can cause lifelong morbidity due to the disconnect that occurs between nerve cells and their cellular targets. Re-establishing these lost connections is the ultimate goal of endogenous regenerative mechanisms, as well as those induced by exogenous manipulations in a laboratory or clinical setting. Reconnection between severed neuronal fibers occurs spontaneously in some invertebrate species and can be induced in mammalian systems. This process, known as axonal fusion, represents a highly efficient means of repair after injury. Recent progress has greatly enhanced our understanding of the molecular control of axonal fusion, demonstrating that the machinery required for the engulfment of apoptotic cells is repurposed to mediate the reconnection between severed axon fragments, which are subsequently merged by fusogen proteins. Here, we review our current understanding of naturally occurring axonal fusion events, as well as those being ectopically produced with the aim of achieving better clinical outcomes.
Collapse
Affiliation(s)
- Brent Neumann
- Neuroscience Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne VIC 3800, Australia.
| | - Casey Linton
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Rosina Giordano-Santini
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Massimo A Hilliard
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia.
| |
Collapse
|
63
|
Geden MJ, Romero SE, Deshmukh M. Apoptosis versus axon pruning: Molecular intersection of two distinct pathways for axon degeneration. Neurosci Res 2018; 139:3-8. [PMID: 30452947 DOI: 10.1016/j.neures.2018.11.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Revised: 11/02/2018] [Accepted: 11/07/2018] [Indexed: 12/16/2022]
Abstract
Neurons are capable of degenerating their axons for the physiological clearance and refinement of unnecessary connections via the programmed degenerative pathways of apoptosis and axon pruning. While both pathways mediate axon degeneration they are however distinct. Whereas in apoptosis the entire neuron, both axons and cell body, degenerates, in the context of axon pruning only the targeted axon segments are selectively degenerated. Interestingly, the molecular pathways mediating axon degeneration in these two contexts have significant mechanistic overlap but also retain distinct differences. In this review, we describe the peripheral neuronal cell culture models used to study the molecular pathways of apoptosis and pruning. We outline what is known about the molecular mechanisms of apoptosis and axon pruning and focus on highlighting the similarities and differences of these two pathways.
Collapse
Affiliation(s)
- Matthew J Geden
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, 27599, USA; Neuroscience Center, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Selena E Romero
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, 27599, USA; Neuroscience Center, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Mohanish Deshmukh
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, 27599, USA; Neuroscience Center, University of North Carolina, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
64
|
Phosphatidylserine is a marker for axonal debris engulfment but its exposure can be decoupled from degeneration. Cell Death Dis 2018; 9:1116. [PMID: 30389906 PMCID: PMC6214901 DOI: 10.1038/s41419-018-1155-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 09/25/2018] [Accepted: 10/08/2018] [Indexed: 02/08/2023]
Abstract
Apoptotic cells expose Phosphatidylserine (PS), that serves as an “eat me” signal for engulfing cells. Previous studies have shown that PS also marks degenerating axonsduring developmental pruning or in response to insults (Wallerian degeneration), but the pathways that control PS exposure on degenerating axons are largely unknown. Here, we used a series of in vitro assays to systematically explore the regulation of PS exposure during axonal degeneration. Our results show that PS exposure is regulated by the upstream activators of axonal pruning and Wallerian degeneration. However, our investigation of signaling further downstream revealed divergence between axon degeneration and PS exposure. Importantly, elevation of the axonal energetic status hindered PS exposure, while inhibition of mitochondrial activity caused PS exposure, without degeneration. Overall, our results suggest that the levels of PS on the outer axonal membrane can be dissociated from the degeneration process and that the axonal energetic status plays a key role in the regulation of PS exposure.
Collapse
|
65
|
Tang HM, Tang HL. Anastasis: recovery from the brink of cell death. ROYAL SOCIETY OPEN SCIENCE 2018; 5:180442. [PMID: 30839720 PMCID: PMC6170572 DOI: 10.1098/rsos.180442] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 08/23/2018] [Indexed: 05/11/2023]
Abstract
Anastasis is a natural cell recovery phenomenon that rescues cells from the brink of death. Programmed cell death such as apoptosis has been traditionally assumed to be an intrinsically irreversible cascade that commits cells to a rapid and massive demolition. Interestingly, recent studies have demonstrated recovery of dying cells even at the late stages generally considered immutable. Here, we examine the evidence for anastasis in cultured cells and in animals, review findings illuminating the potential mechanisms of action, discuss the challenges of studying anastasis and explore new strategies to uncover the function and regulation of anastasis, the identification of which has wide-ranging physiological, pathological and therapeutic implications.
Collapse
Affiliation(s)
- Ho Man Tang
- Institute for Basic Biomedical Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- School of Life Sciences, Chinese University of Hong Kong, Shatin, Hong Kong
| | - Ho Lam Tang
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
66
|
Johnstone AD, Hallett RM, de Léon A, Carturan B, Gibon J, Barker PA. A novel method for quantifying axon degeneration. PLoS One 2018; 13:e0199570. [PMID: 30020957 PMCID: PMC6051587 DOI: 10.1371/journal.pone.0199570] [Citation(s) in RCA: 7] [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: 03/08/2018] [Accepted: 06/08/2018] [Indexed: 11/25/2022] Open
Abstract
Axons normally degenerate during development of the mammalian nervous system, but dysregulation of the same genetically-encoded destructive cellular machinery can destroy crucial structures during adult neurodegenerative diseases. Nerve growth factor (NGF) withdrawal from dorsal root ganglia (DRG) axons is a well-established in vitro experimental model for biochemical and cell biological studies of developmental degeneration. Definitive methods for measuring axon degeneration have been lacking and here we report a novel method of axon degeneration quantification from bulk cultures of DRG that enables objective and automated measurement of axonal density over the entire field of radial axon outgrowth from the ganglion. As proof of principal, this new method, written as an R script called Axoquant 2.0, was used to examine the role of extracellular Ca2+ in the execution of cytoskeletal disassembly during degeneration of NGF-deprived DRG axons. This method can be easily applied to examine degenerative or neuroprotective effects of gene manipulations and pharmacological interventions.
Collapse
Affiliation(s)
- Aaron D. Johnstone
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
- Department of Biology, University of British Columbia Okanagan, Kelowna, British Columbia, Canada
| | - Robin M. Hallett
- The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Andrés de Léon
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
- Department of Biology, University of British Columbia Okanagan, Kelowna, British Columbia, Canada
| | - Bruno Carturan
- Department of Biology, University of British Columbia Okanagan, Kelowna, British Columbia, Canada
| | - Julien Gibon
- Department of Biology, University of British Columbia Okanagan, Kelowna, British Columbia, Canada
| | - Philip A. Barker
- Department of Biology, University of British Columbia Okanagan, Kelowna, British Columbia, Canada
| |
Collapse
|
67
|
Sasaki Y. Metabolic aspects of neuronal degeneration: From a NAD + point of view. Neurosci Res 2018; 139:9-20. [PMID: 30006197 DOI: 10.1016/j.neures.2018.07.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 06/21/2018] [Accepted: 06/22/2018] [Indexed: 12/14/2022]
Abstract
Cellular metabolism maintains the life of cells, allowing energy production required for building cellular constituents and maintaining homeostasis under constantly changing external environments. Neuronal cells maintain their structure and function for the entire life of organisms and the loss of neurons, with limited neurogenesis in adults, directly causes loss of complexity in the neuronal networks. The nervous system organizes the neurons by placing cell bodies containing nuclei of similar types of neurons in discrete regions. Accordingly, axons must travel great distances to connect different types of neurons and peripheral organs. The enormous surface area of neurons makes them high-energy demanding to keep their membrane potential. Distal axon survival is dependent on axonal transport that is another energy demanding process. All of these factors make metabolic stress a potential risk factor for neuronal death and neuronal degeneration often associated with metabolic diseases. This review discusses recent findings on metabolic dysregulations under neuronal degeneration and pathways protecting neurons in these conditions.
Collapse
Affiliation(s)
- Yo Sasaki
- Department of Genetics, Washington University in St. Louis, Couch Biomedical Research Building, 4515 McKinley Ave., Saint Louis, MO, 63110, United States
| |
Collapse
|
68
|
Wang Y, Song M, Song F. Neuronal autophagy and axon degeneration. Cell Mol Life Sci 2018; 75:2389-2406. [PMID: 29675785 PMCID: PMC11105516 DOI: 10.1007/s00018-018-2812-1] [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] [Received: 12/31/2017] [Revised: 03/13/2018] [Accepted: 04/03/2018] [Indexed: 02/08/2023]
Abstract
Axon degeneration is a pathophysiological process of axonal dying and breakdown, which is characterized by several morphological features including the accumulation of axoplasmic organelles, disassembly of microtubules, and fragmentation of the axonal cytoskeleton. Autophagy, a highly conserved lysosomal-degradation machinery responsible for the control of cellular protein quality, is widely believed to be essential for the maintenance of axonal homeostasis in neurons. In recent years, more and more evidence suggests that dysfunctional autophagy is associated with axonal degeneration in many neurodegenerative diseases. Here, we review the core machinery of autophagy in neuronal cells, and provide several major steps that interfere with autophagy flux in neurodegenerative conditions. Furthermore, this review highlights the potential role of neuronal autophagy in axon degeneration, and presents some possible molecular mechanisms by which dysfunctional autophagy leads to axon degeneration in pathological conditions.
Collapse
Affiliation(s)
- Yu Wang
- Department of Toxicology, School of Public Health, Shandong University, 44 Wenhuaxi Road, Jinan, 250012, Shandong, People's Republic of China
| | - Mingxue Song
- School of Public Health, Fujian Medical University, 1 Xueyuan Road, Fuzhou, 350108, Fujian, People's Republic of China
| | - Fuyong Song
- Department of Toxicology, School of Public Health, Shandong University, 44 Wenhuaxi Road, Jinan, 250012, Shandong, People's Republic of China.
| |
Collapse
|
69
|
Shrirao AB, Kung FH, Omelchenko A, Schloss RS, Boustany NN, Zahn JD, Yarmush ML, Firestein BL. Microfluidic platforms for the study of neuronal injury in vitro. Biotechnol Bioeng 2018; 115:815-830. [PMID: 29251352 DOI: 10.1002/bit.26519] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 12/11/2017] [Accepted: 12/15/2017] [Indexed: 12/27/2022]
Abstract
Traumatic brain injury (TBI) affects 5.3 million people in the United States, and there are 12,500 new cases of spinal cord injury (SCI) every year. There is yet a significant need for in vitro models of TBI and SCI in order to understand the biological mechanisms underlying central nervous system (CNS) injury and to identify and test therapeutics to aid in recovery from neuronal injuries. While TBI or SCI studies have been aided with traditional in vivo and in vitro models, the innate limitations in specificity of injury, isolation of neuronal regions, and reproducibility of these models can decrease their usefulness in examining the neurobiology of injury. Microfluidic devices provide several advantages over traditional methods by allowing researchers to (1) examine the effect of injury on specific neural components, (2) fluidically isolate neuronal regions to examine specific effects on subcellular components, and (3) reproducibly create a variety of injuries to model TBI and SCI. These microfluidic devices are adaptable for modeling a wide range of injuries, and in this review, we will examine different methodologies and models recently utilized to examine neuronal injury. Specifically, we will examine vacuum-assisted axotomy, physical injury, chemical injury, and laser-based axotomy. Finally, we will discuss the benefits and downsides to each type of injury model and discuss how researchers can use these parameters to pick a particular microfluidic device to model CNS injury.
Collapse
Affiliation(s)
- Anil B Shrirao
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey
| | - Frank H Kung
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, New Jersey
| | - Anton Omelchenko
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, New Jersey
| | - Rene S Schloss
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey
| | - Nada N Boustany
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey
| | - Jeffrey D Zahn
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey
| | - Martin L Yarmush
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey
| | - Bonnie L Firestein
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, New Jersey
| |
Collapse
|
70
|
Glutamate-induced and NMDA receptor-mediated neurodegeneration entails P2Y1 receptor activation. Cell Death Dis 2018; 9:297. [PMID: 29463792 PMCID: PMC5833818 DOI: 10.1038/s41419-018-0351-1] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 12/28/2017] [Accepted: 01/24/2018] [Indexed: 12/27/2022]
Abstract
Despite the characteristic etiologies and phenotypes, different brain disorders rely on common pathogenic events. Glutamate-induced neurotoxicity is a pathogenic event shared by different brain disorders. Another event occurring in different brain pathological conditions is the increase of the extracellular ATP levels, which is now recognized as a danger and harmful signal in the brain, as heralded by the ability of P2 receptors (P2Rs) to affect a wide range of brain disorders. Yet, how ATP and P2R contribute to neurodegeneration remains poorly defined. For that purpose, we now examined the contribution of extracellular ATP and P2Rs to glutamate-induced neurodegeneration. We found both in vitro and in vivo that ATP/ADP through the activation of P2Y1R contributes to glutamate-induced neuronal death in the rat hippocampus. We found in cultured rat hippocampal neurons that the exposure to glutamate (100 µM) for 30 min triggers a sustained increase of extracellular ATP levels, which contributes to NMDA receptor (NMDAR)-mediated hippocampal neuronal death through the activation of P2Y1R. We also determined that P2Y1R is involved in excitotoxicity in vivo as the blockade of P2Y1R significantly attenuated rat hippocampal neuronal death upon the systemic administration of kainic acid or upon the intrahippocampal injection of quinolinic acid. This contribution of P2Y1R fades with increasing intensity of excitotoxic conditions, which indicates that P2Y1R is not contributing directly to neurodegeneration, rather behaving as a catalyst decreasing the threshold from which glutamate becomes neurotoxic. Moreover, we unraveled that such excitotoxicity process began with an early synaptotoxicity that was also prevented/attenuated by the antagonism of P2Y1R, both in vitro and in vivo. This should rely on the observed glutamate-induced calpain-mediated axonal cytoskeleton damage, most likely favored by a P2Y1R-driven increase of NMDAR-mediated Ca2+ entry selectively in axons. This may constitute a degenerative mechanism shared by different brain diseases, particularly relevant at initial pathogenic stages.
Collapse
|
71
|
Katsuki H, Hijioka M. Intracerebral Hemorrhage as an Axonal Tract Injury Disorder with Inflammatory Reactions. Biol Pharm Bull 2018; 40:564-568. [PMID: 28458342 DOI: 10.1248/bpb.b16-01013] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Intracerebral hemorrhage (ICH) is a neurological disorder frequently accompanied by severe dysfunction. Critical pathogenic events leading to poor prognosis should be identified for the development of novel effective therapies for ICH. Here we focus on the injury of the axonal tract, particularly of the internal capsule, with reference to its contribution to ICH pathology and potential therapeutic interventions in addition to its cellular mechanisms. Studies on human ICH patients and rodent models of ICH suggest that invasion of hematoma into the internal capsule greatly worsens the severity of post-ICH symptoms. A blood-derived protease thrombin may play an important role in the acute phase of axonal tract injury in the internal capsule that includes compromised axonal transport and fragmentation of axonal structures. Several agents such as clioquinol, melatonin and Am80 (a retinoic acid receptor agonist) have been shown to produce therapeutic effects on rodent models of ICH associated with injury of the internal capsule. In the course of examinations on the effect of Am80, we obtained evidence for the involvement of CXCL2, a neutrophil chemotactic factor, in the pathogenesis of ICH. Accordingly, we also refer to the potential roles of infiltrating neutrophils and inflammatory responses in axonal tract injury and resultant neurological dysfunction in ICH.
Collapse
Affiliation(s)
- Hiroshi Katsuki
- Department of Chemico-Pharmacological Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University
| | - Masanori Hijioka
- Department of Chemico-Pharmacological Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University
| |
Collapse
|
72
|
Tang HM, Fung MC, Tang HL. Detecting Anastasis In Vivo by CaspaseTracker Biosensor. J Vis Exp 2018. [PMID: 29443051 DOI: 10.3791/54107] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Anastasis (Greek for "rising to life") is a recently discovered cell recovery phenomenon whereby dying cells can reverse late-stage cell death processes that are generally assumed to be intrinsically irreversible. Promoting anastasis could in principle rescue or preserve injured cells that are difficult to replace such as cardiomyocytes or neurons, thereby facilitating tissue recovery. Conversely, suppressing anastasis in cancer cells, undergoing apoptosis after anti-cancer therapies, may ensure cancer cell death and reduce the chances of recurrence. However, these studies have been hampered by the lack of tools for tracking the fate of cells that undergo anastasis in live animals. The challenge is to identify the cells that have reversed the cell death process despite their morphologically normal appearance after recovery. To overcome this difficulty, we have developed Drosophila and mammalian CaspaseTracker biosensor systems that can identify and permanently track the anastatic cells in vitro or in vivo. Here, we present in vivo protocols for the generation and use of the CaspaseTracker dual biosensor system to detect and track anastasis in Drosophila melanogaster after transient exposure to cell death stimuli. While conventional biosensors and protocols can label cells actively undergoing apoptotic cell death, the CaspaseTracker biosensor can permanently label cells that have recovered after caspase activation - a hallmark of late-stage apoptosis, and simultaneously identify active apoptotic processes. This biosensor can also track the recovery of the cells that attempted other forms of cell death that directly or indirectly involved caspase activity. Therefore, this protocol enables us to continuously track the fate of these cells and their progeny, facilitating future studies of the biological functions, molecular mechanisms, physiological and pathological consequences, and therapeutic implications of anastasis. We also discuss the appropriate controls to distinguish cells that undergo anastasis from those that display non-apoptotic caspase activity in vivo.
Collapse
Affiliation(s)
- Ho Man Tang
- Institute for Basic Biomedical Sciences, Johns Hopkins University School of Medicine; School of Life Sciences, Chinese University of Hong Kong;
| | - Ming Chiu Fung
- School of Life Sciences, Chinese University of Hong Kong;
| | - Ho Lam Tang
- Department of Neurosurgery, Johns Hopkins University School of Medicine;
| |
Collapse
|
73
|
Abstract
SARM1 is a key regulator of axonal degeneration. However, SARM1 mechanism of action is not clear. In this issue of Neuron, Essuman et al. (2017) reveal an intrinsic NADase activity in the SARM1-TIR domain that is required for axonal degeneration.
Collapse
|
74
|
Feinberg K, Kolaj A, Wu C, Grinshtein N, Krieger JR, Moran MF, Rubin LL, Miller FD, Kaplan DR. A neuroprotective agent that inactivates prodegenerative TrkA and preserves mitochondria. J Cell Biol 2017; 216:3655-3675. [PMID: 28877995 PMCID: PMC5674898 DOI: 10.1083/jcb.201705085] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 07/26/2017] [Accepted: 08/01/2017] [Indexed: 12/15/2022] Open
Abstract
The pan-kinase inhibitor foretinib is identified as a potent suppressor of sympathetic, sensory, and motor neuron axon degeneration, acting in part by inhibiting the activity of the unliganded TrkA/nerve growth factor receptor and by preserving mitochondria in die-back and Wallerian degeneration models. Axon degeneration is an early event and pathological in neurodegenerative conditions and nerve injuries. To discover agents that suppress neuronal death and axonal degeneration, we performed drug screens on primary rodent neurons and identified the pan-kinase inhibitor foretinib, which potently rescued sympathetic, sensory, and motor wt and SOD1 mutant neurons from trophic factor withdrawal-induced degeneration. By using primary sympathetic neurons grown in mass cultures and Campenot chambers, we show that foretinib protected neurons by suppressing both known degenerative pathways and a new pathway involving unliganded TrkA and transcriptional regulation of the proapoptotic BH3 family members BimEL, Harakiri,and Puma, culminating in preservation of mitochondria in the degenerative setting. Foretinib delayed chemotherapy-induced and Wallerian axonal degeneration in culture by preventing axotomy-induced local energy deficit and preserving mitochondria, and peripheral Wallerian degeneration in vivo. These findings identify a new axon degeneration pathway and a potentially clinically useful therapeutic drug.
Collapse
Affiliation(s)
- Konstantin Feinberg
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON, Canada
| | - Adelaida Kolaj
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON, Canada.,Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Chen Wu
- Department of Stem Cell and Regenerative Biology and Harvard Stem Cell Institute, Harvard University, Cambridge, MA
| | - Natalie Grinshtein
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON, Canada
| | - Jonathan R Krieger
- Program in Cell Biology, Hospital for Sick Children, Toronto, ON, Canada
| | - Michael F Moran
- Program in Cell Biology, Hospital for Sick Children, Toronto, ON, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Lee L Rubin
- Department of Stem Cell and Regenerative Biology and Harvard Stem Cell Institute, Harvard University, Cambridge, MA
| | - Freda D Miller
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON, Canada .,Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada.,Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - David R Kaplan
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON, Canada .,Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
75
|
Salvadores N, Sanhueza M, Manque P, Court FA. Axonal Degeneration during Aging and Its Functional Role in Neurodegenerative Disorders. Front Neurosci 2017; 11:451. [PMID: 28928628 PMCID: PMC5591337 DOI: 10.3389/fnins.2017.00451] [Citation(s) in RCA: 121] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 07/25/2017] [Indexed: 12/11/2022] Open
Abstract
Aging constitutes the main risk factor for the development of neurodegenerative diseases. This represents a major health issue worldwide that is only expected to escalate due to the ever-increasing life expectancy of the population. Interestingly, axonal degeneration, which occurs at early stages of neurodegenerative disorders (ND) such as Alzheimer's disease, Amyotrophic lateral sclerosis, and Parkinson's disease, also takes place as a consequence of normal aging. Moreover, the alteration of several cellular processes such as proteostasis, response to cellular stress and mitochondrial homeostasis, which have been described to occur in the aging brain, can also contribute to axonal pathology. Compelling evidence indicate that the degeneration of axons precedes clinical symptoms in NDs and occurs before cell body loss, constituting an early event in the pathological process and providing a potential therapeutic target to treat neurodegeneration before neuronal cell death. Although, normal aging and the development of neurodegeneration are two processes that are closely linked, the molecular basis of the switch that triggers the transition from healthy aging to neurodegeneration remains unrevealed. In this review we discuss the potential role of axonal degeneration in this transition and provide a detailed overview of the literature and current advances in the molecular understanding of the cellular changes that occur during aging that promote axonal degeneration and then discuss this in the context of ND.
Collapse
Affiliation(s)
- Natalia Salvadores
- Center for Integrative Biology, Faculty of Sciences, Universidad MayorSantiago, Chile.,Fondap Geroscience Center for Brain Health and MetabolismSantiago, Chile
| | - Mario Sanhueza
- Center for Integrative Biology, Faculty of Sciences, Universidad MayorSantiago, Chile.,Fondap Geroscience Center for Brain Health and MetabolismSantiago, Chile
| | - Patricio Manque
- Center for Integrative Biology, Faculty of Sciences, Universidad MayorSantiago, Chile
| | - Felipe A Court
- Center for Integrative Biology, Faculty of Sciences, Universidad MayorSantiago, Chile.,Fondap Geroscience Center for Brain Health and MetabolismSantiago, Chile
| |
Collapse
|
76
|
TMEM184b Promotes Axon Degeneration and Neuromuscular Junction Maintenance. J Neurosci 2017; 36:4681-9. [PMID: 27122027 DOI: 10.1523/jneurosci.2893-15.2016] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 03/07/2016] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED Complex nervous systems achieve proper connectivity during development and must maintain these connections throughout life. The processes of axon and synaptic maintenance and axon degeneration after injury are jointly controlled by a number of proteins within neurons, including ubiquitin ligases and mitogen activated protein kinases. However, our understanding of these molecular cascades is incomplete. Here we describe the phenotype resulting from mutation of TMEM184b, a protein identified in a screen for axon degeneration mediators. TMEM184b is highly expressed in the mouse nervous system and is found in recycling endosomes in neuronal cell bodies and axons. Disruption of TMEM184b expression results in prolonged maintenance of peripheral axons following nerve injury, demonstrating a role for TMEM184b in axon degeneration. In contrast to this protective phenotype in axons, uninjured mutant mice have anatomical and functional impairments in the peripheral nervous system. Loss of TMEM184b causes swellings at neuromuscular junctions that become more numerous with age, demonstrating that TMEM184b is critical for the maintenance of synaptic architecture. These swellings contain abnormal multivesicular structures similar to those seen in patients with neurodegenerative disorders. Mutant animals also show abnormal sensory terminal morphology. TMEM184b mutant animals are deficient on the inverted screen test, illustrating a role for TMEM184b in sensory-motor function. Overall, we have identified an important function for TMEM184b in peripheral nerve terminal structure, function, and the axon degeneration pathway. SIGNIFICANCE STATEMENT Our work has identified both neuroprotective and neurodegenerative roles for a previously undescribed protein, TMEM184b. TMEM184b mutation causes delayed axon degeneration following peripheral nerve injury, indicating that it participates in the degeneration process. Simultaneously, TMEM184b mutation causes progressive structural abnormalities at neuromuscular synapses and swellings within sensory terminals, and animals with this mutation display profound weakness. Thus, TMEM184b is necessary for normal peripheral nerve terminal morphology and maintenance. Loss of TMEM184b results in accumulation of autophagosomal structures in vivo, fitting with emerging studies that have linked autophagy disruption and neurological disease. Our work recognizes TMEM184b as a new player in the maintenance of the nervous system.
Collapse
|
77
|
Lee YJ, Cintora P, Arikkath J, Akinsola O, Kandel M, Popescu G, Best-Popescu C. Quantitative assessment of neural outgrowth using spatial light interference microscopy. JOURNAL OF BIOMEDICAL OPTICS 2017; 22:66015. [PMID: 28655053 PMCID: PMC5482290 DOI: 10.1117/1.jbo.22.6.066015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 05/30/2017] [Indexed: 05/12/2023]
Abstract
Optimal growth as well as branching of axons and dendrites is critical for the nervous system function. Neuritic length, arborization, and growth rate determine the innervation properties of neurons and define each cell’s computational capability. Thus, to investigate the nervous system function, we need to develop methods and instrumentation techniques capable of quantifying various aspects of neural network formation: neuron process extension, retraction, stability, and branching. During the last three decades, fluorescence microscopy has yielded enormous advances in our understanding of neurobiology. While fluorescent markers provide valuable specificity to imaging, photobleaching, and photoxicity often limit the duration of the investigation. Here, we used spatial light interference microscopy (SLIM) to measure quantitatively neurite outgrowth as a function of cell confluence. Because it is label-free and nondestructive, SLIM allows for long-term investigation over many hours. We found that neurons exhibit a higher growth rate of neurite length in low-confluence versus medium- and high-confluence conditions. We believe this methodology will aid investigators in performing unbiased, nondestructive analysis of morphometric neuronal parameters.
Collapse
Affiliation(s)
- Young Jae Lee
- University of Illinois at Urbana-Champaign, Department of Bioengineering, Cellular Neuroscience and Imaging Laboratory, Urbana, Illinois, United States
| | - Pati Cintora
- University of Illinois at Urbana-Champaign, Department of Bioengineering, Cellular Neuroscience and Imaging Laboratory, Urbana, Illinois, United States
| | - Jyothi Arikkath
- University of Nebraska Medical Center, Munroe-Meyer Institute, Omaha, Nebraska, United States
| | - Olaoluwa Akinsola
- University of Illinois at Urbana-Champaign, Beckman Institute for Advanced Science and Technology, Department of Electrical and Computer Engineering, Quantitative Light Imaging Laboratory, Urbana, Illinois, United States
| | - Mikhail Kandel
- University of Illinois at Urbana-Champaign, Beckman Institute for Advanced Science and Technology, Department of Electrical and Computer Engineering, Quantitative Light Imaging Laboratory, Urbana, Illinois, United States
| | - Gabriel Popescu
- University of Illinois at Urbana-Champaign, Beckman Institute for Advanced Science and Technology, Department of Electrical and Computer Engineering, Quantitative Light Imaging Laboratory, Urbana, Illinois, United States
| | - Catherine Best-Popescu
- University of Illinois at Urbana-Champaign, Department of Bioengineering, Cellular Neuroscience and Imaging Laboratory, Urbana, Illinois, United States
| |
Collapse
|
78
|
Hao Y, Collins C. Intrinsic mechanisms for axon regeneration: insights from injured axons in Drosophila. Curr Opin Genet Dev 2017; 44:84-91. [PMID: 28232273 PMCID: PMC5447494 DOI: 10.1016/j.gde.2017.01.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 01/17/2017] [Accepted: 01/26/2017] [Indexed: 12/18/2022]
Abstract
Axonal damage and loss are common and negative consequences of neuronal injuries, and also occur in some neurodegenerative diseases. For neurons to have a chance to repair their connections, they need to survive the damage, initiate new axonal growth, and ultimately establish new synaptic connections. This review discusses how recent work in Drosophila models have informed our understanding of the cellular pathways used by neurons to respond to axonal injuries. Similarly to mammalian neurons, Drosophila neurons appear to be more limited in their capacity regrow (regenerate) damaged axons in the central nervous system, but can undergo axonal regeneration to varying extents in the peripheral nervous system. Conserved cellular pathways are activated by axonal injury via mechanisms that are specific to axons but not dendrites, and new unanticipated inhibitors of axon regeneration can be identified via genetic screening. These findings, made predominantly via genetic and live imaging methods in Drosophila, emphasize the utility of this model organism for the identification and study of basic cellular mechanisms used for neuronal repair.
Collapse
Affiliation(s)
- Yan Hao
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109-1048, USA
| | - Catherine Collins
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109-1048, USA.
| |
Collapse
|
79
|
Chen YJ, Nabavizadeh SA, Vossough A, Kumar S, Loevner LA, Mohan S. Wallerian Degeneration Beyond the Corticospinal Tracts: Conventional and Advanced MRI Findings. J Neuroimaging 2017; 27:272-280. [PMID: 28072502 DOI: 10.1111/jon.12404] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 10/04/2016] [Accepted: 10/11/2016] [Indexed: 12/21/2022] Open
Abstract
Wallerian degeneration (WD) is defined as progressive anterograde disintegration of axons and accompanying demyelination after an injury to the proximal axon or cell body. Since the 1980s and 1990s, conventional magnetic resonance imaging (MRI) sequences have been shown to be sensitive to changes of WD in the subacute to chronic phases. More recently, advanced MRI techniques, such as diffusion-weighted imaging (DWI) and diffusion tensor imaging (DTI), have demonstrated some of earliest changes attributed to acute WD, typically on the order of days. In addition, there is increasing evidence on the value of advanced MRI techniques in providing important prognostic information related to WD. This article reviews the utility of conventional and advanced MRI techniques for assessing WD, by focusing not only on the corticospinal tract but also other neural tracts less commonly thought of, including corticopontocerebellar tract, dentate-rubro-olivary pathway, posterior column of the spinal cord, corpus callosum, limbic circuit, and optic pathway. The basic anatomy of these neural pathways will be discussed, followed by a comprehensive review of existing literature supported by instructive clinical examples. The goal of this review is for readers to become more familiar with both conventional and advanced MRI findings of WD involving important neural pathways, as well as to illustrate increasing utility of advanced MRI techniques in providing important prognostic information for various pathologies.
Collapse
Affiliation(s)
- Yin Jie Chen
- Division of Neuroradiology, Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Seyed Ali Nabavizadeh
- Division of Neuroradiology, Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Arastoo Vossough
- Department of Radiology, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Sunil Kumar
- Department of Radiodiagnosis, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Laurie A Loevner
- Division of Neuroradiology, Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Suyash Mohan
- Division of Neuroradiology, Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
80
|
Wang S, Wang S, Asgar J, Joseph J, Ro JY, Wei F, Campbell JN, Chung MK. Ca 2+ and calpain mediate capsaicin-induced ablation of axonal terminals expressing transient receptor potential vanilloid 1. J Biol Chem 2017; 292:8291-8303. [PMID: 28360106 DOI: 10.1074/jbc.m117.778290] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 03/28/2017] [Indexed: 01/01/2023] Open
Abstract
Capsaicin is an ingredient in spicy peppers that produces burning pain by activating transient receptor potential vanilloid 1 (TRPV1), a Ca2+-permeable ion channel in nociceptors. Capsaicin has also been used as an analgesic, and its topical administration is approved for the treatment of certain pain conditions. The mechanisms underlying capsaicin-induced analgesia likely involve reversible ablation of nociceptor terminals. However, the mechanisms underlying these effects are not well understood. To visualize TRPV1-lineage axons, a genetically engineered mouse model was used in which a fluorophore is expressed under the TRPV1 promoter. Using a combination of these TRPV1-lineage reporter mice and primary afferent cultures, we monitored capsaicin-induced effects on afferent terminals in real time. We found that Ca2+ influx through TRPV1 is necessary for capsaicin-induced ablation of nociceptive terminals. Although capsaicin-induced mitochondrial Ca2+ uptake was TRPV1-dependent, dissipation of the mitochondrial membrane potential, inhibition of the mitochondrial transition permeability pore, and scavengers of reactive oxygen species did not attenuate capsaicin-induced ablation. In contrast, MDL28170, an inhibitor of the Ca2+-dependent protease calpain, diminished ablation. Furthermore, overexpression of calpastatin, an endogenous inhibitor of calpain, or knockdown of calpain 2 also decreased ablation. Quantitative assessment of TRPV1-lineage afferents in the epidermis of the hind paws of the reporter mice showed that EGTA and MDL28170 diminished capsaicin-induced ablation. Moreover, MDL28170 prevented capsaicin-induced thermal hypoalgesia. These results suggest that TRPV1/Ca2+/calpain-dependent signaling plays a dominant role in capsaicin-induced ablation of nociceptive terminals and further our understanding of the molecular mechanisms underlying the effects of capsaicin on nociceptors.
Collapse
Affiliation(s)
- Sheng Wang
- Department of Neural and Pain Sciences, School of Dentistry, Program in Neuroscience, Center to Advance Chronic Pain Research, University of Maryland, Baltimore, Maryland 21201
| | - Sen Wang
- Department of Neural and Pain Sciences, School of Dentistry, Program in Neuroscience, Center to Advance Chronic Pain Research, University of Maryland, Baltimore, Maryland 21201
| | - Jamila Asgar
- Department of Neural and Pain Sciences, School of Dentistry, Program in Neuroscience, Center to Advance Chronic Pain Research, University of Maryland, Baltimore, Maryland 21201
| | - John Joseph
- Department of Neural and Pain Sciences, School of Dentistry, Program in Neuroscience, Center to Advance Chronic Pain Research, University of Maryland, Baltimore, Maryland 21201
| | - Jin Y Ro
- Department of Neural and Pain Sciences, School of Dentistry, Program in Neuroscience, Center to Advance Chronic Pain Research, University of Maryland, Baltimore, Maryland 21201
| | - Feng Wei
- Department of Neural and Pain Sciences, School of Dentistry, Program in Neuroscience, Center to Advance Chronic Pain Research, University of Maryland, Baltimore, Maryland 21201
| | | | - Man-Kyo Chung
- Department of Neural and Pain Sciences, School of Dentistry, Program in Neuroscience, Center to Advance Chronic Pain Research, University of Maryland, Baltimore, Maryland 21201.
| |
Collapse
|
81
|
Wrobel MR, Sundararaghavan HG. Positive and negative cues for modulating neurite dynamics and receptor expression. Biomed Mater 2017; 12:025016. [DOI: 10.1088/1748-605x/aa61d1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
82
|
Li LS, Lu YL, Nie J, Xu YY, Zhang W, Yang WJ, Gong QH, Lu YF, Lu Y, Shi JS. Dendrobium nobile Lindl alkaloid, a novel autophagy inducer, protects against axonal degeneration induced by Aβ 25-35 in hippocampus neurons in vitro. CNS Neurosci Ther 2017; 23:329-340. [PMID: 28261990 DOI: 10.1111/cns.12678] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 01/12/2017] [Accepted: 01/16/2017] [Indexed: 12/19/2022] Open
Abstract
AIMS Axonal degeneration is a pathological symbol in the early stage of Alzheimer's disease (AD), which can be triggered by amyloid-β (Aβ) peptide deposition. Growing evidence indicates that deficit of autophagy eventually leads to the axonal degeneration. Our previous studies have shown that Dendrobium nobile Lindl alkaloid (DNLA) had protective effect on neuron impairment in vivo and in vitro; however, the underlying mechanisms is still unclear. METHODS We exposed cultured hippocampus neurons to Aβ25-35 to investigate the effect of DNLA in vitro. Axonal degeneration was evaluated by immunofluorescence staining and MTT assay. Neurons overexpressing GFP-LC3B were used to measure the formation of autophagosome. Autophagosome-lysosome fusion, the lysosomal pH, and cathepsin activity were assessed to reflect autophagy process. Proteins of interest were analyzed by Western blot. RESULTS DNLA pretreatment significantly inhibited axonal degeneration induced by Aβ25-35 peptide in vitro. Further studies revealed DNLA treatment increased autophagic flux through promoting formation and degradation of autophagosome in hippocampus neurons. Moreover, enhancement of autophagic flux was responsible for the protective effects of DNLA on axonal degeneration. CONCLUSIONS DNLA prevents Aβ25-35 -induced axonal degeneration via activation of autophagy process and could be a novel therapeutic target.
Collapse
Affiliation(s)
- Li-Sheng Li
- Department of Chemistry, Basic Medical Faculty, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Pharmacology, Key Lab of Basic Pharmacology of Education Ministry, Zunyi Medical University, Zunyi, Guizhou, China
| | - Yan-Liu Lu
- Department of Pharmacology, Key Lab of Basic Pharmacology of Education Ministry, Zunyi Medical University, Zunyi, Guizhou, China
| | - Jing Nie
- Department of Pharmacology, Key Lab of Basic Pharmacology of Education Ministry, Zunyi Medical University, Zunyi, Guizhou, China
| | - Yun-Yan Xu
- Department of Pharmacology, Key Lab of Basic Pharmacology of Education Ministry, Zunyi Medical University, Zunyi, Guizhou, China
| | - Wei Zhang
- Department of Pharmacology, Key Lab of Basic Pharmacology of Education Ministry, Zunyi Medical University, Zunyi, Guizhou, China
| | - Wen-Jin Yang
- Department of Pharmacology, Key Lab of Basic Pharmacology of Education Ministry, Zunyi Medical University, Zunyi, Guizhou, China
| | - Qi-Hai Gong
- Department of Pharmacology, Key Lab of Basic Pharmacology of Education Ministry, Zunyi Medical University, Zunyi, Guizhou, China
| | - Yuan-Fu Lu
- Department of Pharmacology, Key Lab of Basic Pharmacology of Education Ministry, Zunyi Medical University, Zunyi, Guizhou, China
| | - Yang Lu
- Department of Chemistry, Basic Medical Faculty, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing-Shan Shi
- Department of Pharmacology, Key Lab of Basic Pharmacology of Education Ministry, Zunyi Medical University, Zunyi, Guizhou, China
| |
Collapse
|
83
|
Molecular pathogenesis of peripheral neuropathies: insights from Drosophila models. Curr Opin Genet Dev 2017; 44:61-73. [PMID: 28213160 DOI: 10.1016/j.gde.2017.01.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 01/10/2017] [Accepted: 01/26/2017] [Indexed: 01/18/2023]
Abstract
Peripheral neuropathies are characterized by degeneration of peripheral motor, sensory and/or autonomic axons, leading to progressive distal muscle weakness, sensory deficits and/or autonomic dysfunction. Acquired peripheral neuropathies, e.g., as a side effect of chemotherapy, are distinguished from inherited peripheral neuropathies (IPNs). Drosophila models for chemotherapy-induced peripheral neuropathy and several IPNs have provided novel insight into the molecular mechanisms underlying axonal degeneration. Forward genetic screens have predictive value for discovery of human IPN genes, and the pathogenicity of novel mutations in known IPN genes can be evaluated in Drosophila. Future screens for genes and compounds that modify Drosophila IPN phenotypes promise to make valuable contributions to unraveling the molecular pathogenesis and identification of therapeutic targets for these incurable diseases.
Collapse
|
84
|
Holland SM, Thomas GM. Roles of palmitoylation in axon growth, degeneration and regeneration. J Neurosci Res 2017; 95:1528-1539. [PMID: 28150429 DOI: 10.1002/jnr.24003] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 11/09/2016] [Accepted: 11/28/2016] [Indexed: 12/11/2022]
Abstract
The protein-lipid modification palmitoylation plays important roles in neurons, but most attention has focused on roles of this modification in the regulation of mature pre- and post-synapses. However, exciting recent findings suggest that palmitoylation is also critical for both the growth and integrity of neuronal axons and plays previously unappreciated roles in conveying axonal anterograde and retrograde signals. Here we review these emerging roles for palmitoylation in the regulation of axons in health and disease. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Sabrina M Holland
- Shriners Hospitals Pediatric Research Center (Center for Neurorehabilitation and Neural Repair)
| | - Gareth M Thomas
- Shriners Hospitals Pediatric Research Center (Center for Neurorehabilitation and Neural Repair).,Department of Anatomy and Cell Biology, Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA, 19140
| |
Collapse
|
85
|
Lai YW, Chu SY, Wei JY, Cheng CY, Li JC, Chen PL, Chen CH, Yu HH. Drosophila microRNA-34 Impairs Axon Pruning of Mushroom Body γ Neurons by Downregulating the Expression of Ecdysone Receptor. Sci Rep 2016; 6:39141. [PMID: 28008974 PMCID: PMC5180235 DOI: 10.1038/srep39141] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 11/17/2016] [Indexed: 11/25/2022] Open
Abstract
MicroRNA-34 (miR-34) is crucial for preventing chronic large-scale neurite degeneration in the aged brain of Drosophila melanogaster. Here we investigated the role of miR-34 in two other types of large-scale axon degeneration in Drosophila: axotomy-induced axon degeneration in olfactory sensory neurons (OSNs) and developmentally related axon pruning in mushroom body (MB) neurons. Ectopically overexpressed miR-34 did not inhibit axon degeneration in OSNs following axotomy, whereas ectopically overexpressed miR-34 in differentiated MB neurons impaired γ axon pruning. Intriguingly, the miR-34-induced γ axon pruning defect resulted from downregulating the expression of ecdysone receptor B1 (EcR-B1) in differentiated MB γ neurons. Notably, the separate overexpression of EcR-B1 or a transforming growth factor- β receptor Baboon, whose activation can upregulate the EcR-B1 expression, in MB neurons rescued the miR-34-induced γ axon pruning phenotype. Future investigations of miR-34 targets that regulate the expression of EcR-B1 in MB γ neurons are warranted to elucidate pathways that regulate axon pruning, and to provide insight into mechanisms that control large-scale axon degeneration in the nervous system.
Collapse
Affiliation(s)
- Yen-Wei Lai
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan.,Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli County, Taiwan.,Institute of Molecular and Cellular Biology, College of Life Science, National Taiwan University, Taipei, Taiwan
| | - Sao-Yu Chu
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Jia-Yi Wei
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Chu-Ya Cheng
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli County, Taiwan
| | - Jian-Chiuan Li
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli County, Taiwan
| | - Po-Lin Chen
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli County, Taiwan.,Institute of Molecular and Cellular Biology, College of Life Science, National Taiwan University, Taipei, Taiwan
| | - Chun-Hong Chen
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli County, Taiwan.,Institute of Molecular and Cellular Biology, College of Life Science, National Taiwan University, Taipei, Taiwan
| | - Hung-Hsiang Yu
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
86
|
Maor-Nof M, Romi E, Sar Shalom H, Ulisse V, Raanan C, Nof A, Leshkowitz D, Lang R, Yaron A. Axonal Degeneration Is Regulated by a Transcriptional Program that Coordinates Expression of Pro- and Anti-degenerative Factors. Neuron 2016; 92:991-1006. [DOI: 10.1016/j.neuron.2016.10.061] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 06/15/2016] [Accepted: 10/20/2016] [Indexed: 12/27/2022]
|
87
|
Monitoring peripheral nerve degeneration in ALS by label-free stimulated Raman scattering imaging. Nat Commun 2016; 7:13283. [PMID: 27796305 PMCID: PMC5095598 DOI: 10.1038/ncomms13283] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 09/19/2016] [Indexed: 01/02/2023] Open
Abstract
The study of amyotrophic lateral sclerosis (ALS) and potential interventions would be facilitated if motor axon degeneration could be more readily visualized. Here we demonstrate that stimulated Raman scattering (SRS) microscopy could be used to sensitively monitor peripheral nerve degeneration in ALS mouse models and ALS autopsy materials. Three-dimensional imaging of pre-symptomatic SOD1 mouse models and data processing by a correlation-based algorithm revealed that significant degeneration of peripheral nerves could be detected coincidentally with the earliest detectable signs of muscle denervation and preceded physiologically measurable motor function decline. We also found that peripheral degeneration was an early event in FUS as well as C9ORF72 repeat expansion models of ALS, and that serial imaging allowed long-term observation of disease progression and drug effects in living animals. Our study demonstrates that SRS imaging is a sensitive and quantitative means of measuring disease progression, greatly facilitating future studies of disease mechanisms and candidate therapeutics.
Collapse
|
88
|
Calpain-dependent truncated form of TrkB-FL increases in neurodegenerative processes. Mol Cell Neurosci 2016; 75:81-92. [PMID: 27449758 DOI: 10.1016/j.mcn.2016.07.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 06/24/2016] [Accepted: 07/19/2016] [Indexed: 11/24/2022] Open
Abstract
Recent findings indicate that the mechanisms that drive reshaping of the nervous system are aberrantly activated in epilepsy and several neurodegenerative diseases. The recurrent seizures in epilepsy, particularly in the condition called status epilepticus, can cause permanent neurological damage, resulting in cognitive dysfunction and other serious neurological conditions. In this study, we used an in vitro model of status epilepticus to examine the role of calpain in the degeneration of hippocampal neurons. We grew neurons on a culture system that allowed studying the dendritic and axonal domains separately from the cell bodies. We found that a recently characterized calpain substrate, the neurotrophin receptor TrkB, is cleaved in the dendritic and axonal domain of neurons committed to die, and this constitutes an early step in the neuronal degeneration process. While the full-length TrkB (TrkB-FL) levels decreased, the truncated form of TrkB (Tc TrkB-FL) concurrently increased, leading to a TrkB-FL/Tc TrkB-FL imbalance, which is thought to be causally linked to neurodegeneration. We further show that the treatment with N-acetyl-Leu-Leu-norleucinal, a specific calpain activity blocker, fully protects the neuronal processes from degeneration, prevents the TrkB-FL/Tc TrkB-FL imbalance, and provides full neuroprotection. Moreover, the use of the TrkB antagonist ANA 12 at the time when the levels of TrkB-FL were significantly decreased, totally blocked neuronal death, suggesting that Tc TrkB-FL may have a role in neuronal death. These results indicate that the imbalance of these neurotrophins receptors plays a key role in neurite degeneration induced by seizures.
Collapse
|
89
|
Brettle M, Patel S, Fath T. Tropomyosins in the healthy and diseased nervous system. Brain Res Bull 2016; 126:311-323. [PMID: 27298153 DOI: 10.1016/j.brainresbull.2016.06.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 06/09/2016] [Accepted: 06/10/2016] [Indexed: 12/25/2022]
Abstract
Regulation of the actin cytoskeleton is dependent on a plethora of actin-associated proteins in all eukaryotic cells. The family of tropomyosins plays a key role in controlling the function of several of these actin-associated proteins and their access to actin filaments. In order to understand the regulation of the actin cytoskeleton in highly dynamic subcellular compartments of neurons such as growth cones of developing neurons and the synaptic compartment of mature neurons, it is pivotal to decipher the functional role of tropomyosins in the nervous system. In this review, we will discuss the current understanding and recent findings on the regulation of the actin cytoskeleton by tropomyosins and potential implication that this has for the dysregulation of the actin cytoskeleton in neurological diseases.
Collapse
Affiliation(s)
- Merryn Brettle
- Neurodegeneration and Repair Unit, School of Medical Sciences, University of New South Wales, 2052 Sydney, New South Wales, Australia
| | - Shrujna Patel
- Neurodegeneration and Repair Unit, School of Medical Sciences, University of New South Wales, 2052 Sydney, New South Wales, Australia
| | - Thomas Fath
- Neurodegeneration and Repair Unit, School of Medical Sciences, University of New South Wales, 2052 Sydney, New South Wales, Australia.
| |
Collapse
|
90
|
Chang B, Quan Q, Lu S, Wang Y, Peng J. Molecular mechanisms in the initiation phase of Wallerian degeneration. Eur J Neurosci 2016; 44:2040-8. [PMID: 27062141 DOI: 10.1111/ejn.13250] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 04/06/2016] [Accepted: 04/06/2016] [Indexed: 12/20/2022]
Abstract
Axonal degeneration is an early hallmark of nerve injury and many neurodegenerative diseases. The discovery of the Wallerian degeneration slow mutant mouse, in which axonal degeneration is delayed, revealed that Wallerian degeneration is an active progress and thereby illuminated the mechanisms underlying axonal degeneration. Nicotinamide mononucleotide adenylyltransferase 2 and sterile alpha and armadillo motif-containing protein 1 play essential roles in the maintenance of axon integrity by regulating the level of nicotinamide adenine dinucleotide, which seems to be the key molecule involved in the maintenance of axonal health. However, the function of nicotinamide mononucleotide remains debatable, and we discuss two apparently conflicting roles of nicotinamide mononucleotide in Wallerian degeneration. In this article, we focus on the roles of these molecules in the initiation phase of Wallerian degeneration to improve our understanding of the mechanisms underpinning this phenomenon.
Collapse
Affiliation(s)
- Biao Chang
- Institute of Orthopedics, General Hospital of People's Liberation Army, 28th Fuxing Road, Beijing, China.,Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Beijing, China.,Key Laboratory of Musculoskeletal Trauma & War Injuries, People's Liberation Army, Beijing, China
| | - Qi Quan
- Institute of Orthopedics, General Hospital of People's Liberation Army, 28th Fuxing Road, Beijing, China.,Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Beijing, China.,Key Laboratory of Musculoskeletal Trauma & War Injuries, People's Liberation Army, Beijing, China
| | - Shibi Lu
- Institute of Orthopedics, General Hospital of People's Liberation Army, 28th Fuxing Road, Beijing, China.,Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Beijing, China.,Key Laboratory of Musculoskeletal Trauma & War Injuries, People's Liberation Army, Beijing, China
| | - Yu Wang
- Institute of Orthopedics, General Hospital of People's Liberation Army, 28th Fuxing Road, Beijing, China.,Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Beijing, China.,Key Laboratory of Musculoskeletal Trauma & War Injuries, People's Liberation Army, Beijing, China.,The Neural Regeneration Co-innovation Center of Jiangsu Province, Nantong, China
| | - Jiang Peng
- Institute of Orthopedics, General Hospital of People's Liberation Army, 28th Fuxing Road, Beijing, China.,Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Beijing, China.,Key Laboratory of Musculoskeletal Trauma & War Injuries, People's Liberation Army, Beijing, China.,The Neural Regeneration Co-innovation Center of Jiangsu Province, Nantong, China
| |
Collapse
|
91
|
Axon degeneration: context defines distinct pathways. Curr Opin Neurobiol 2016; 39:108-15. [PMID: 27197022 DOI: 10.1016/j.conb.2016.05.002] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 05/02/2016] [Accepted: 05/03/2016] [Indexed: 12/29/2022]
Abstract
Axon degeneration is an essential part of development, plasticity, and injury response and has been primarily studied in mammalian models in three contexts: 1) Axotomy-induced Wallerian degeneration, 2) Apoptosis-induced axon degeneration (axon apoptosis), and 3) Axon pruning. These three contexts dictate engagement of distinct pathways for axon degeneration. Recent advances have identified the importance of SARM1, NMNATs, NAD+ depletion, and MAPK signaling in axotomy-induced Wallerian degeneration. Interestingly, apoptosis-induced axon degeneration and axon pruning have many shared mechanisms both in signaling (e.g. DLK, JNKs, GSK3α/β) and execution (e.g. Puma, Bax, caspase-9, caspase-3). However, the specific mechanisms by which caspases are activated during apoptosis versus pruning appear distinct, with apoptosis requiring Apaf-1 but not caspase-6 while pruning requires caspase-6 but not Apaf-1.
Collapse
|
92
|
Abstract
Immature neural circuits form excessive synaptic connections that are later refined through pruning of exuberant branches. In this issue, Bornstein et al. identify a role for JNK signaling in selective axon elimination through disassembly of cell adhesion complexes.
Collapse
Affiliation(s)
- Jonathan E Farley
- Department of Neurobiology/HHMI, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Marc R Freeman
- Department of Neurobiology/HHMI, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
93
|
Human Umbilical Tissue-Derived Cells Promote Synapse Formation and Neurite Outgrowth via Thrombospondin Family Proteins. J Neurosci 2016; 35:15649-65. [PMID: 26609158 DOI: 10.1523/jneurosci.1364-15.2015] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
UNLABELLED Cell therapy demonstrates great potential for the treatment of neurological disorders. Human umbilical tissue-derived cells (hUTCs) were previously shown to have protective and regenerative effects in animal models of stroke and retinal degeneration, but the underlying therapeutic mechanisms are unknown. Because synaptic dysfunction, synapse loss, degeneration of neuronal processes, and neuronal death are hallmarks of neurological diseases and retinal degenerations, we tested whether hUTCs contribute to tissue repair and regeneration by stimulating synapse formation, neurite outgrowth, and neuronal survival. To do so, we used a purified rat retinal ganglion cell culture system and found that hUTCs secrete factors that strongly promote excitatory synaptic connectivity and enhance neuronal survival. Additionally, we demonstrated that hUTCs support neurite outgrowth under normal culture conditions and in the presence of the growth-inhibitory proteins chondroitin sulfate proteoglycan, myelin basic protein, or Nogo-A (reticulon 4). Furthermore, through biochemical fractionation and pharmacology, we identified the major hUTC-secreted synaptogenic factors as the thrombospondin family proteins (TSPs), TSP1, TSP2, and TSP4. Silencing TSP expression in hUTCs, using small RNA interference, eliminated both the synaptogenic function of these cells and their ability to promote neurite outgrowth. However, the majority of the prosurvival functions of hUTC-conditioned media was spared after TSP knockdown, indicating that hUTCs secrete additional neurotrophic factors. Together, our findings demonstrate that hUTCs affect multiple aspects of neuronal health and connectivity through secreted factors, and each of these paracrine effects may individually contribute to the therapeutic function of these cells. SIGNIFICANCE STATEMENT Human umbilical tissue-derived cells (hUTC) are currently under clinical investigation for the treatment of geographic atrophy secondary to age-related macular degeneration. These cells show great promise for the treatment of neurological disorders; however, the therapeutic effects of these cells on CNS neurons are not fully understood. Here we provide compelling evidence that hUTCs secrete multiple factors that work synergistically to enhance synapse formation and function, and support neuronal growth and survival. Moreover, we identified thrombospondins (TSPs) as the hUTC-secreted factors that mediate the synaptogenic and growth-promoting functions of these cells. Our findings highlight novel paracrine effects of hUTC on CNS neuron health and connectivity and begin to unravel potential therapeutic mechanisms by which these cells elicit their effects.
Collapse
|
94
|
Abstract
The assembly of functional neural circuits requires the combined action of progressive and regressive events. Regressive events encompass a variety of inhibitory developmental processes, including axon and dendrite pruning, which facilitate the removal of exuberant neuronal connections. Most axon pruning involves the removal of axons that had already made synaptic connections; thus, axon pruning is tightly associated with synapse elimination. In many instances, these developmental processes are regulated by the interplay between neurons and glial cells that act instructively during neural remodeling. Owing to the importance of axon and dendritic pruning, these remodeling events require precise spatial and temporal control, and this is achieved by a range of distinct molecular mechanisms. Disruption of these mechanisms results in abnormal pruning, which has been linked to brain dysfunction. Therefore, understanding the mechanisms of axon and dendritic pruning will be instrumental in advancing our knowledge of neural disease and mental disorders.
Collapse
Affiliation(s)
- Martin M Riccomagno
- Department of Cell Biology and Neuroscience, University of California, Riverside, California 92521;
| | - Alex L Kolodkin
- Solomon H. Snyder Department of Neuroscience, Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205;
| |
Collapse
|
95
|
Suzuki K, Lovera M, Schmachtenberg O, Couve E. Axonal Degeneration in Dental Pulp Precedes Human Primary Teeth Exfoliation. J Dent Res 2015; 94:1446-53. [PMID: 26149320 DOI: 10.1177/0022034515593055] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The dental pulp in human primary teeth is densely innervated by a plethora of nerve endings at the coronal pulp-dentin interface. This study analyzed how the physiological root resorption (PRR) process affects dental pulp innervation before exfoliation of primary teeth. Forty-four primary canine teeth, classified into 3 defined PRR stages (early, middle, and advanced) were fixed and demineralized. Longitudinal cryosections of each tooth were stained for immunohistochemical and quantitative analysis of dental pulp nerve fibers and associated components with confocal and electron microscopy. During PRR, axonal degeneration was prominent and progressive in a Wallerian-like scheme, comprising nerve fiber bundles and nerve endings within the coronal and root pulp. Neurofilament fragmentation increased significantly during PRR progression and was accompanied by myelin degradation and a progressive loss of myelinated axons. Myelin sheath degradation involved activation of autophagic activity by Schwann cells to remove myelin debris. These cells expressed a sequence of responses comprising dedifferentiation, proliferative activity, GAP-43 overexpression, and Büngner band formation. During the advanced PRR stage, increased immune cell recruitment within the dental pulp and major histocompatibility complex (MHC) class II upregulation by Schwann cells characterized an inflammatory condition associated with the denervation process in preexfoliative primary teeth. The ensuing loss of dental pulp axons is likely to be responsible for the progressive reduction of sensory function of the dental pulp during preexfoliative stages.
Collapse
Affiliation(s)
- K Suzuki
- Clínica de Odontología Pediátrica y del Adolescente, Universidad de Valparaíso, Valparaíso, Chile Facultad de Odontología, Universidad de Valparaíso, Valparaíso, Chile
| | - M Lovera
- Facultad de Odontología, Universidad de Valparaíso, Valparaíso, Chile
| | - O Schmachtenberg
- Centro Interdisciplinario de Neurociencia de Valparaíso (CINV), Universidad de Valparaíso, Valparaíso, Chile Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - E Couve
- Centro Interdisciplinario de Neurociencia de Valparaíso (CINV), Universidad de Valparaíso, Valparaíso, Chile Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile Instituto de Biología, Laboratorio de Microscopía Electrónica, Universidad de Valparaíso, Valparaíso, Chile
| |
Collapse
|
96
|
Rodriguez M, Rodriguez-Sabate C, Morales I, Sanchez A, Sabate M. Parkinson's disease as a result of aging. Aging Cell 2015; 14:293-308. [PMID: 25677794 PMCID: PMC4406659 DOI: 10.1111/acel.12312] [Citation(s) in RCA: 142] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/16/2014] [Indexed: 12/15/2022] Open
Abstract
It is generally considered that Parkinson's disease is induced by specific agents that degenerate a clearly defined population of dopaminergic neurons. Data commented in this review suggest that this assumption is not as clear as is often thought and that aging may be critical for Parkinson's disease. Neurons degenerating in Parkinson's disease also degenerate in normal aging, and the different agents involved in the etiology of this illness are also involved in aging. Senescence is a wider phenomenon affecting cells all over the body, whereas Parkinson's disease seems to be restricted to certain brain centers and cell populations. However, reviewed data suggest that Parkinson's disease may be a local expression of aging on cell populations which, by their characteristics (high number of synaptic terminals and mitochondria, unmyelinated axons, etc.), are highly vulnerable to the agents promoting aging. The development of new knowledge about Parkinson's disease could be accelerated if the research on aging and Parkinson's disease were planned together, and the perspective provided by gerontology gains relevance in this field.
Collapse
Affiliation(s)
- Manuel Rodriguez
- Laboratory of Neurobiology and Experimental Neurology, Department of Physiology, Faculty of Medicine, University of La LagunaLa Laguna, Spain
- Center for Networked Biomedical Research in Neurodegenerative Diseases (CIBERNED)La Laguna, Spain
| | - Clara Rodriguez-Sabate
- Center for Networked Biomedical Research in Neurodegenerative Diseases (CIBERNED)La Laguna, Spain
| | - Ingrid Morales
- Laboratory of Neurobiology and Experimental Neurology, Department of Physiology, Faculty of Medicine, University of La LagunaLa Laguna, Spain
- Center for Networked Biomedical Research in Neurodegenerative Diseases (CIBERNED)La Laguna, Spain
| | - Alberto Sanchez
- Laboratory of Neurobiology and Experimental Neurology, Department of Physiology, Faculty of Medicine, University of La LagunaLa Laguna, Spain
| | - Magdalena Sabate
- Rehabilitation Service, Department of Pharmacology and Physical Medicine, Faculty of Medicine, University of La LagunaLa Laguna, Spain
| |
Collapse
|
97
|
Gilley J, Orsomando G, Nascimento-Ferreira I, Coleman MP. Absence of SARM1 rescues development and survival of NMNAT2-deficient axons. Cell Rep 2015; 10:1974-81. [PMID: 25818290 PMCID: PMC4386025 DOI: 10.1016/j.celrep.2015.02.060] [Citation(s) in RCA: 148] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Revised: 12/23/2014] [Accepted: 02/24/2015] [Indexed: 11/01/2022] Open
Abstract
SARM1 function and nicotinamide mononucleotide adenylyltransferase 2 (NMNAT2) loss both promote axon degeneration, but their relative relationship in the process is unknown. Here, we show that NMNAT2 loss and resultant changes to NMNAT metabolites occur in injured SARM1-deficient axons despite their delayed degeneration and that axon degeneration specifically induced by NMNAT2 depletion requires SARM1. Strikingly, SARM1 deficiency also corrects axon outgrowth in mice lacking NMNAT2, independently of NMNAT metabolites, preventing perinatal lethality. Furthermore, NAMPT inhibition partially restores outgrowth of NMNAT2-deficient axons, suggesting that the NMNAT substrate, NMN, contributes to this phenotype. NMNAT2-depletion-dependent degeneration of established axons and restricted extension of developing axons are thus both SARM1 dependent, and SARM1 acts either downstream of NMNAT2 loss and NMN accumulation in a linear pathway or in a parallel branch of a convergent pathway. Understanding the pathway will help establish relationships with other modulators of axon survival and facilitate the development of effective therapies for axonopathies.
Collapse
Affiliation(s)
- Jonathan Gilley
- Signalling Programme, Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Giuseppe Orsomando
- Department of Clinical Sciences (DISCO), Section of Biochemistry, Polytechnic University of Marche, Via Ranieri 67, Ancona 60131, Italy
| | | | - Michael P Coleman
- Signalling Programme, Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK.
| |
Collapse
|
98
|
Klistorner A, Vootakuru N, Wang C, Yiannikas C, Graham SL, Parratt J, Garrick R, Levin N, Masters L, Lagopoulos J, Barnett MH. Decoding diffusivity in multiple sclerosis: analysis of optic radiation lesional and non-lesional white matter. PLoS One 2015; 10:e0122114. [PMID: 25807541 PMCID: PMC4373765 DOI: 10.1371/journal.pone.0122114] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 02/17/2015] [Indexed: 12/19/2022] Open
Abstract
Objectives Diffusion tensor imaging (DTI) has been suggested as a new promising tool in MS that may provide greater pathological specificity than conventional MRI, helping, therefore, to elucidate disease pathogenesis and monitor therapeutic efficacy. However, the pathological substrates that underpin alterations in brain tissue diffusivity are not yet fully delineated. Tract-specific DTI analysis has previously been proposed in an attempt to alleviate this problem. Here, we extended this approach by segmenting a single tract into areas bound by seemingly similar pathological processes, which may better delineate the potential association between DTI metrics and underlying tissue damage. Method Several compartments were segmented in optic radiation (OR) of 50 relapsing-remitting MS patients including T2 lesions, proximal and distal parts of fibers transected by lesion and fibers with no discernable pathology throughout the entire length of the OR. Results Asymmetry analysis between lesional and non-lesional fibers demonstrated a marked increase in Radial Diffusivity (RD), which was topographically limited to focal T2 lesions and potentially relates to the lesional myelin loss. A relative elevation of Axial Diffusivity (AD) in the distal part of the lesional fibers was observed in a distribution consistent with Wallerian degeneration, while diffusivity in the proximal portion of transected axons remained normal. A moderate, but significant elevation of RD in OR non-lesional fibers was strongly associated with the global (but not local) T2 lesion burden and is probably related to microscopic demyelination undetected by conventional MRI. Conclusion This study highlights the utility of the compartmentalization approach in elucidating the pathological substrates of diffusivity and demonstrates the presence of tissue-specific patterns of altered diffusivity in MS, providing further evidence that DTI is a sensitive marker of tissue damage in both lesions and NAWM. Our results suggest that, at least within the OR, parallel and perpendicular diffusivities are affected by tissue restructuring related to distinct pathological processes.
Collapse
Affiliation(s)
- Alexander Klistorner
- Department of Ophthalmology, Save Sight Institute, University of Sydney, Sydney, Australia
- Australian School of Advanced Medicine, Macquarie University, Sydney, NSW, Australia
- * E-mail:
| | | | - Chenyu Wang
- Brain and Mind Research Institute, University of Sydney, Sydney, NSW, Australia
| | | | - Stuart L. Graham
- Australian School of Advanced Medicine, Macquarie University, Sydney, NSW, Australia
| | | | | | - Netta Levin
- Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Lynette Masters
- Brain and Mind Research Institute, University of Sydney, Sydney, NSW, Australia
| | - Jim Lagopoulos
- Brain and Mind Research Institute, University of Sydney, Sydney, NSW, Australia
| | - Michael H. Barnett
- Brain and Mind Research Institute, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
99
|
Kanamori T, Yoshino J, Yasunaga KI, Dairyo Y, Emoto K. Local endocytosis triggers dendritic thinning and pruning in Drosophila sensory neurons. Nat Commun 2015; 6:6515. [DOI: 10.1038/ncomms7515] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Accepted: 02/04/2015] [Indexed: 12/15/2022] Open
|
100
|
In vivo CaspaseTracker biosensor system for detecting anastasis and non-apoptotic caspase activity. Sci Rep 2015; 5:9015. [PMID: 25757939 PMCID: PMC4355673 DOI: 10.1038/srep09015] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Accepted: 02/16/2015] [Indexed: 11/17/2022] Open
Abstract
The discovery that mammalian cells can survive late-stage apoptosis challenges the general assumption that active caspases are markers of impending death. However, tools have not been available to track healthy cells that have experienced caspase activity at any time in the past. Therefore, to determine if cells in whole animals can undergo reversal of apoptosis, known as anastasis, we developed a dual color CaspaseTracker system for Drosophila to identify cells with ongoing or past caspase activity. Transient exposure of healthy females to environmental stresses such as cold shock or starvation activated the CaspaseTracker coincident with caspase activity and apoptotic morphologies in multiple cell types of developing egg chambers. Importantly, when stressed flies were returned to normal conditions, morphologically healthy egg chambers and new progeny flies were labeled by the biosensor, suggesting functional recovery from apoptotic caspase activation. In striking contrast to developing egg chambers, which lack basal caspase biosensor activation under normal conditions, many adult tissues of normal healthy flies exhibit robust caspase biosensor activity in a portion of cells, including neurons. The widespread persistence of CaspaseTracker-positivity implies that healthy cells utilize active caspases for non-apoptotic physiological functions during and after normal development.
Collapse
|