1
|
oboma YI, Eberechukwu GC. Alteration in Oxidative Stress Enzymes and Proteins by Datura metel Stramonium Hydroethanolic Root Extract Promotes Hippocampus and Cerebral Cortex Neurons Damage in Adult Sprague Dawley Rats. Subst Abuse Rehabil 2025; 16:13-22. [PMID: 39882249 PMCID: PMC11776506 DOI: 10.2147/sar.s490392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 01/21/2025] [Indexed: 01/31/2025] Open
Abstract
Background Datura metel is reported to induce hallucinations and mental disorders. Objective This study investigates the neurotoxic effects of Datura metel stramonium hydroethanolic root extract on the hippocampus and cerebral cortex of adult rats using biochemical, histological and immunohistochemical techniques. Methodology Twenty five adult rats were assigned to 5 groups (n = 5 each). Group A - negative control, group B (lead positive control). Groups C, D, and E exposed to 150 mg/kg, 300 mg/kg and 600 mg/kg body weight of extracts for 14 days once daily, respectively. Histology, biochemistry and immunohistochemical techniques were used to study cell injury in the brain tissue. Results Biochemical alterations were observed in superoxide dismutase (SOD), Malondialdehyde (MDA), Glutathionine S-transferase (GST) and Catalase among the experimental groups. Catalase was statistically significant at P<0.05. Histology reveals neurons damage, depletion and vacuolation. NFP and NSE were over expressed in the experimental groups. Discussion Oral administration of Datura metel root extracts at high concentration alters the antioxidant enzymes activity and body weight. Extracts cause cortex and hippocampus neurotoxicity through heightened oxidative stress. Conclusion Datura metel root extract is a neurotoxic agent and causes depopulation of hippocampus and cerebral cortex neurons. The use of this plant should be highly regulated to reduce neuropathies associated with consumption.
Collapse
Affiliation(s)
- Yibala Ibor oboma
- Department of Medical Laboratory Science, Faculty of Basic Medical Sciences, College of Health Sciences, Niger Delta University Wilberforce Island Bayelsa State, Amassoma, Nigeria
- Department of Medical Laboratory Science, School of Allied Health Sciences, Kampala International University, Ishaka Municipal, Uganda
| | - God’sman C Eberechukwu
- Department of Medical Laboratory Science, Faculty of Basic Medical Sciences, College of Health Sciences, Niger Delta University Wilberforce Island Bayelsa State, Amassoma, Nigeria
- Department of Histopathology and Cytopathology, Faculty of Medical Laboratory Science, Federal University, Otuoke, Bayelsa State, Nigeria
| |
Collapse
|
2
|
Merkulyeva N, Mikhalkin A. Transient expression of heavy-chain neurofilaments in the perigeniculate nucleus of cats. Brain Struct Funct 2024; 229:489-495. [PMID: 38265459 DOI: 10.1007/s00429-023-02752-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 12/12/2023] [Indexed: 01/25/2024]
Abstract
The perigeniculate nucleus (PGN) is a visual part of the thalamic reticular nucleus modulating the information transfer between the lateral geniculate nucleus and the visual cortex. This study focused on the postnatal development of the PGN in cats, using the SMI-32 antibody, which recognizes non-phosphorylated heavy-chain neurofilaments responsible for neuronal structural maturation and is also used as a marker for motion processing, or Y, stream. We questioned whether transient neuronal populations exist in the PGN and can they possibly be related to the Y processing stream. We uncovered a transient, robust SMI-32 staining in the PGN of kittens aged 0-34 days with the significant decline in the cellular density of labeled cells in older animals. According to the double-labeling, in all examined age groups, perigeniculate SMI-32-immunopositive cells are part of the main parvalbumin-positive population. The maximal cellular density of the double-stained cells appeared in animals aged 10-28 days. We also revealed that the most significant growth of perigeniculate cells's soma occurred at three postnatal weeks. The possible link of our data to the development of the Y visual processing stream and to the heterogeneity of the perigeniculate neuronal population is also discussed.
Collapse
Affiliation(s)
- Natalia Merkulyeva
- Pavlov Institute of Physiology RAS, Makarov Nab., 6, Saint-Petersburg, Russia, 199034.
| | - Aleksandr Mikhalkin
- Pavlov Institute of Physiology RAS, Makarov Nab., 6, Saint-Petersburg, Russia, 199034
| |
Collapse
|
3
|
Kleinschmidt H, Xu C, Bai L. Using Synthetic DNA Libraries to Investigate Chromatin and Gene Regulation. Chromosoma 2023; 132:167-189. [PMID: 37184694 PMCID: PMC10542970 DOI: 10.1007/s00412-023-00796-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 04/25/2023] [Accepted: 04/26/2023] [Indexed: 05/16/2023]
Abstract
Despite the recent explosion in genome-wide studies in chromatin and gene regulation, we are still far from extracting a set of genetic rules that can predict the function of the regulatory genome. One major reason for this deficiency is that gene regulation is a multi-layered process that involves an enormous variable space, which cannot be fully explored using native genomes. This problem can be partially solved by introducing synthetic DNA libraries into cells, a method that can test the regulatory roles of thousands to millions of sequences with limited variables. Here, we review recent applications of this method to study transcription factor (TF) binding, nucleosome positioning, and transcriptional activity. We discuss the design principles, experimental procedures, and major findings from these studies and compare the pros and cons of different approaches.
Collapse
Affiliation(s)
- Holly Kleinschmidt
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA, 16802, USA
- Center for Eukaryotic Gene Regulation, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Cheng Xu
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA, 16802, USA
- Center for Eukaryotic Gene Regulation, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Lu Bai
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA, 16802, USA.
- Center for Eukaryotic Gene Regulation, The Pennsylvania State University, University Park, PA, 16802, USA.
- Department of Physics, The Pennsylvania State University, University Park, PA, 16802, USA.
| |
Collapse
|
4
|
Hour NL, Cabana T, Pflieger JF. Transient expression of NF200 by fibers in the nasal septum and rostral telencephalon of developing opossums (Monodelphis domestica). Anat Rec (Hoboken) 2023; 306:879-888. [PMID: 36056623 DOI: 10.1002/ar.25057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 07/14/2022] [Accepted: 08/11/2022] [Indexed: 11/10/2022]
Abstract
Marsupials are born very immature and crawl on their mother's belly to attach to teats. Sensory information is required to guide the newborn and to induce attachment to the teat. Olfaction has been classically proposed to influence neonatal behaviors, but recent studies suggest that the central olfactory structures are too immature to account for them. In the newborn opossum, we previously described a fascicle of nerve fibers expressing neurofilament-200 (NF200, a marker of fiber maturity) from the olfactory bulbs to the rostral telencephalon. The course of these fibers is compatible with that of the terminal nerve that, during development, is characterized by the presence of neurons synthetizing gonadotropin hormones (GnRH). To evaluate if these fibers are related to the terminal nerve and if they play a role in precocious behaviors in opossums, we used immunohistochemistry against NF200 and GnRH. The results show that NF200-labeled fibers are present between P0 and P11, but do not reach much further caudally than the septal region. Only a few NF200-labeled fibers were found near the olfactory and vomeronasal epitheliums and they did not penetrate the olfactory bulbs. NF200-labeled fibers follow the same path as fibers labeled for GnRH. In contrast to the latter, NF200-labeled fibers are no longer visible at P15. These results suggest that these fibers are neither from the olfactory nor from the vomeronasal nerves but may be part of the terminal nerve. Their limited caudal extension does not support a role in the sensorimotor behaviors of the newborn opossum.
Collapse
|
5
|
Del Tredici K, Braak H. Neuropathology and neuroanatomy of TDP-43 amyotrophic lateral sclerosis. Curr Opin Neurol 2022; 35:660-671. [PMID: 36069419 DOI: 10.1097/wco.0000000000001098] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
PURPOSE OF REVIEW Intracellular inclusions consisting of the abnormal TDP-43 protein and its nucleocytoplasmic mislocalization in selected cell types are hallmark pathological features of sALS. Descriptive (histological, morphological), anatomical, and molecular studies all have improved our understanding of the neuropathology of sporadic amyotrophic lateral sclerosis (sALS). This review highlights some of the latest developments in the field. RECENT FINDINGS Increasing evidence exists from experimental models for the prion-like nature of abnormal TDP-43, including a strain-effect, and with the help of neuroimaging-based studies, for spreading of disease along corticofugal connectivities in sALS. Progress has also been made with respect to finding and establishing reliable biomarkers (neurofilament levels, diffusor tensor imaging). SUMMARY The latest findings may help to elucidate the preclinical phase of sALS and to define possible mechanisms for delaying or halting disease development and progression.
Collapse
Affiliation(s)
- Kelly Del Tredici
- Clinical Neuroanatomy Section, Department of Neurology, Center for Biomedical Research, University of Ulm, Ulm, Germany
| | | |
Collapse
|
6
|
Will Cannabigerol Trigger Neuroregeneration after a Spinal Cord Injury? An In Vitro Answer from NSC-34 Scratch-Injured Cells Transcriptome. Pharmaceuticals (Basel) 2022; 15:ph15020117. [PMID: 35215230 PMCID: PMC8875351 DOI: 10.3390/ph15020117] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/12/2022] [Accepted: 01/17/2022] [Indexed: 02/07/2023] Open
Abstract
Spinal cord injury affects the lives of millions of people around the world, often causing disability and, in unfortunate circumstances, death. Rehabilitation can partly improve outcomes and only a small percentage of patients, typically the least injured, can hope to return to normal living conditions. Cannabis sativa is gaining more and more interest in recent years, even though its beneficial properties have been known for thousands of years. Cannabigerol (CBG), extracted from C. sativa, is defined as the “mother of all cannabinoids” and its properties range from anti-inflammatory to antioxidant and neuroprotection. Using NSC-34 cells to model spinal cord injury in vitro, our work evaluated the properties of CBG treatments in motor neuron regeneration. While pre-treatment can modulate oxidative stress and increase antioxidant enzyme genes, such as Tnx1, decreasing Nos1 post-treatment seems to induce regeneration genes by triggering different pathways, such as Gap43 via p53 acetylation by Ep300 and Ddit3 and Xbp1 via Bdnf signaling, along with cytoskeletal remodeling signaling genes Nrp1 and Map1b. Our results indicate CBG as a phytocompound worth further investigation in the field of neuronal regeneration.
Collapse
|
7
|
Feliciano CM, Wu K, Watry HL, Marley CBE, Ramadoss GN, Ghanim HY, Liu AZ, Zholudeva LV, McDevitt TC, Saporta MA, Conklin BR, Judge LM. Allele-Specific Gene Editing Rescues Pathology in a Human Model of Charcot-Marie-Tooth Disease Type 2E. Front Cell Dev Biol 2021; 9:723023. [PMID: 34485306 PMCID: PMC8415563 DOI: 10.3389/fcell.2021.723023] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 07/22/2021] [Indexed: 12/26/2022] Open
Abstract
Many neuromuscular disorders are caused by dominant missense mutations that lead to dominant-negative or gain-of-function pathology. This category of disease is challenging to address via drug treatment or gene augmentation therapy because these strategies may not eliminate the effects of the mutant protein or RNA. Thus, effective treatments are severely lacking for these dominant diseases, which often cause severe disability or death. The targeted inactivation of dominant disease alleles by gene editing is a promising approach with the potential to completely remove the cause of pathology with a single treatment. Here, we demonstrate that allele-specific CRISPR gene editing in a human model of axonal Charcot-Marie-Tooth (CMT) disease rescues pathology caused by a dominant missense mutation in the neurofilament light chain gene (NEFL, CMT type 2E). We utilized a rapid and efficient method for generating spinal motor neurons from human induced pluripotent stem cells (iPSCs) derived from a patient with CMT2E. Diseased motor neurons recapitulated known pathologic phenotypes at early time points of differentiation, including aberrant accumulation of neurofilament light chain protein in neuronal cell bodies. We selectively inactivated the disease NEFL allele in patient iPSCs using Cas9 enzymes to introduce a frameshift at the pathogenic N98S mutation. Motor neurons carrying this allele-specific frameshift demonstrated an amelioration of the disease phenotype comparable to that seen in an isogenic control with precise correction of the mutation. Our results validate allele-specific gene editing as a therapeutic approach for CMT2E and as a promising strategy to silence dominant mutations in any gene for which heterozygous loss-of-function is well tolerated. This highlights the potential for gene editing as a therapy for currently untreatable dominant neurologic diseases.
Collapse
Affiliation(s)
- Carissa M. Feliciano
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, United States
- Gladstone Institutes, San Francisco, CA, United States
| | - Kenneth Wu
- Gladstone Institutes, San Francisco, CA, United States
| | | | - Chiara B. E. Marley
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, United States
- Gladstone Institutes, San Francisco, CA, United States
| | - Gokul N. Ramadoss
- Gladstone Institutes, San Francisco, CA, United States
- Biomedical Sciences Ph.D. Program, University of California, San Francisco, San Francisco, CA, United States
| | | | - Angela Z. Liu
- Gladstone Institutes, San Francisco, CA, United States
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, United States
| | | | - Todd C. McDevitt
- Gladstone Institutes, San Francisco, CA, United States
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, United States
| | - Mario A. Saporta
- Department of Neurology, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Bruce R. Conklin
- Gladstone Institutes, San Francisco, CA, United States
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, United States
- Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
- Innovative Genomics Institute, Berkeley, CA, United States
| | - Luke M. Judge
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, United States
- Gladstone Institutes, San Francisco, CA, United States
| |
Collapse
|
8
|
Quintá HR. Intraspinal Administration of Netrin-1 Promotes Locomotor Recovery after Complete Spinal Cord Transection. J Neurotrauma 2021; 38:2084-2102. [PMID: 33599152 DOI: 10.1089/neu.2020.7571] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Complete spinal cord lesions interrupt the connection of all axonal projections with their neuronal targets below and above the lesion site. In particular, the interruption of connections with the neurons at lumbar segments after thoracic injuries impairs voluntary body control below the injury. The failure of spontaneous regrowth of transected axons across the lesion prevents the reconnection and reinnervation of the neuronal targets. At present, the only treatment in humans that has proven to promote some degree of locomotor recovery is physical therapy. The success of these strategies, however, depends greatly on the type of lesion and the level of preservation of neural tissue in the spinal cord after injury. That is the reason it is key to design strategies to promote axonal regrowth and neuronal reconnection. Here, we test the use of a developmental axon guidance molecule as a biological agent to promote axonal regrowth, axonal reconnection, and recovery of locomotor activity after spinal cord injury (SCI). This molecule, netrin-1, guides the growth of the corticospinal tract (CST) during the development of the central nervous system. To assess the potential of this molecule, we used a model of complete spinal cord transection in rats, at thoracic level 10-11. We show that in situ delivery of netrin-1 at the epicenter of the lesion: (1) promotes regrowth of CST through the lesion and prevents CST dieback, (2) promotes synaptic reconnection of regenerated motor and sensory axons, and (3) preserves the polymerization of the neurofilaments in the sciatic nerve axons. These anatomical findings correlate with a significant recovery of locomotor function. Our work identifies netrin-1 as a biological agent with the capacity to promote the functional repair and recovery of locomotor function after SCI. These findings support the use of netrin-1 as a therapeutic intervention to be tested in humans.
Collapse
Affiliation(s)
- Héctor R Quintá
- Consejo Nacional de Investigaciones Científicas y Técnicas-CONICET, Buenos Aires, Argentina
- Laboratorio de Medicina Experimental "Dr. Jorge E. Toblli," Hospital Alemán. CABA, Buenos Aires, Argentina
| |
Collapse
|
9
|
Khan MI, Hasan F, Mahmud KAHA, Adnan A. Viscoelastic Response of Neurofilaments: An Atomistic Simulation Approach. Biomolecules 2021; 11:biom11040540. [PMID: 33917073 PMCID: PMC8067762 DOI: 10.3390/biom11040540] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 03/29/2021] [Accepted: 04/05/2021] [Indexed: 12/05/2022] Open
Abstract
Existent literature has limitations regarding the mechanical behavior of axonal cytoskeletal components in a high strain rate scenario, which is mainly due to limitations regarding the structure of some components such as tau protein and neurofilaments (NF). This study performs molecular dynamics (MD) simulations on NFs to extract their strain rate-dependent behavior. It is found that they are highly stretchable and show multiple stages of unfolding. Furthermore, NFs show high tensile stiffness. Also, viscoelastic modeling shows that they correspond to simplified viscoelastic models. This study effectively enhances the existent axonal models focusing on axonal injury.
Collapse
|
10
|
Bomont P. The dazzling rise of neurofilaments: Physiological functions and roles as biomarkers. Curr Opin Cell Biol 2021; 68:181-191. [PMID: 33454158 DOI: 10.1016/j.ceb.2020.10.011] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 10/13/2020] [Accepted: 10/19/2020] [Indexed: 12/15/2022]
Abstract
In the last two years, neurofilaments (NFs) have become one of the most blazing topics in clinical neuroscience. NFs are major cytoskeletal constituents of neurons, can be detected in body fluids, and have recently emerged as universal biomarkers of neuronal injury and neurological diseases. This review will examine the evolving landscape of NFs, from their specific cellular functions within neurons to their broad clinical value as biomarkers. Particular attention will be given to the dynamic nature of the NF network and its novel roles in microtubule regulation, neurotransmission, and nanomedicine. Building from the initial evidence of causative mutations in NF genes in Charcot-Marie-Tooth diseases, the latest advances at the frontiers of basic and clinical sciences have expanded the scope and relevance of NFs for human health remarkably and have poised to fuel innovation in cell biology and neuroscience.
Collapse
Affiliation(s)
- Pascale Bomont
- ERC team, INMG, INSERM U1217, CNRS UMR5310, University of Lyon 1, University of Lyon, Lyon, France.
| |
Collapse
|
11
|
The Arg/N-degron pathway targets transcription factors and regulates specific genes. Proc Natl Acad Sci U S A 2020; 117:31094-31104. [PMID: 33229537 DOI: 10.1073/pnas.2020124117] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The Arg/N-degron pathway targets proteins for degradation by recognizing their N-terminal or internal degrons. Our previous work produced double-knockout (2-KO) HEK293T human cell lines that lacked the functionally overlapping UBR1 and UBR2 E3 ubiquitin ligases of the Arg/N-degron pathway. Here, we studied these cells in conjunction with RNA-sequencing, mass spectrometry (MS), and split-ubiquitin binding assays. 1) Some mRNAs, such as those encoding lactate transporter MCT2 and β-adrenergic receptor ADRB2, are strongly (∼20-fold) up-regulated in 2-KO cells, whereas other mRNAs, including those encoding MAGEA6 (a regulator of ubiquitin ligases) and LCP1 (an actin-binding protein), are completely repressed in 2-KO cells, in contrast to wild-type cells. 2) Glucocorticoid receptor (GR), an immunity-modulating transcription factor (TF), is up-regulated in 2-KO cells and also physically binds to UBR1, strongly suggesting that GR is a physiological substrate of the Arg/N-degron pathway. 3) PREP1, another TF, was also found to bind to UBR1. 4) MS-based analyses identified ∼160 proteins whose levels were increased or decreased by more than 2-fold in 2-KO cells. For example, the homeodomain TF DACH1 and the neurofilament subunits NF-L (NFEL) and NF-M (NFEM) were expressed in wild-type cells but were virtually absent in 2-KO cells. 5) The disappearance of some proteins in 2-KO cells took place despite up-regulation of their mRNAs, strongly suggesting that the Arg/N-degron pathway can also modulate translation of specific mRNAs. In sum, this multifunctional proteolytic system has emerged as a regulator of mammalian gene expression, in part through conditional targeting of TFs that include ATF3, GR, and PREP1.
Collapse
|
12
|
Prokop A. Cytoskeletal organization of axons in vertebrates and invertebrates. J Cell Biol 2020; 219:e201912081. [PMID: 32369543 PMCID: PMC7337489 DOI: 10.1083/jcb.201912081] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 04/13/2020] [Accepted: 04/14/2020] [Indexed: 12/11/2022] Open
Abstract
The maintenance of axons for the lifetime of an organism requires an axonal cytoskeleton that is robust but also flexible to adapt to mechanical challenges and to support plastic changes of axon morphology. Furthermore, cytoskeletal organization has to adapt to axons of dramatically different dimensions, and to their compartment-specific requirements in the axon initial segment, in the axon shaft, at synapses or in growth cones. To understand how the cytoskeleton caters to these different demands, this review summarizes five decades of electron microscopic studies. It focuses on the organization of microtubules and neurofilaments in axon shafts in both vertebrate and invertebrate neurons, as well as the axon initial segments of vertebrate motor- and interneurons. Findings from these ultrastructural studies are being interpreted here on the basis of our contemporary molecular understanding. They strongly suggest that axon architecture in animals as diverse as arthropods and vertebrates is dependent on loosely cross-linked bundles of microtubules running all along axons, with only minor roles played by neurofilaments.
Collapse
Affiliation(s)
- Andreas Prokop
- School of Biology, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| |
Collapse
|
13
|
Rodemer W, Gallo G, Selzer ME. Mechanisms of Axon Elongation Following CNS Injury: What Is Happening at the Axon Tip? Front Cell Neurosci 2020; 14:177. [PMID: 32719586 PMCID: PMC7347967 DOI: 10.3389/fncel.2020.00177] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 05/22/2020] [Indexed: 12/12/2022] Open
Abstract
After an injury to the central nervous system (CNS), functional recovery is limited by the inability of severed axons to regenerate and form functional connections with appropriate target neurons beyond the injury. Despite tremendous advances in our understanding of the mechanisms of axon growth, and of the inhibitory factors in the injured CNS that prevent it, disappointingly little progress has been made in restoring function to human patients with CNS injuries, such as spinal cord injury (SCI), through regenerative therapies. Clearly, the large number of overlapping neuron-intrinsic and -extrinsic growth-inhibitory factors attenuates the benefit of neutralizing any one target. More daunting is the distances human axons would have to regenerate to reach some threshold number of target neurons, e.g., those that occupy one complete spinal segment, compared to the distances required in most experimental models, such as mice and rats. However, the difficulties inherent in studying mechanisms of axon regeneration in the mature CNS in vivo have caused researchers to rely heavily on extrapolation from studies of axon regeneration in peripheral nerve, or of growth cone-mediated axon development in vitro and in vivo. Unfortunately, evidence from several animal models, including the transected lamprey spinal cord, has suggested important differences between regeneration of mature CNS axons and growth of axons in peripheral nerve, or during embryonic development. Specifically, long-distance regeneration of severed axons may not involve the actin-myosin molecular motors that guide embryonic growth cones in developing axons. Rather, non-growth cone-mediated axon elongation may be required to propel injured axons in the mature CNS. If so, it may be necessary to use other experimental models to promote regeneration that is sufficient to contact a critical number of target neurons distal to a CNS lesion. This review examines the cytoskeletal underpinnings of axon growth, focusing on the elongating axon tip, to gain insights into how CNS axons respond to injury, and how this might affect the development of regenerative therapies for SCI and other CNS injuries.
Collapse
Affiliation(s)
- William Rodemer
- Shriners Hospitals Pediatric Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Gianluca Gallo
- Shriners Hospitals Pediatric Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States.,Department of Anatomy and Cell Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Michael E Selzer
- Shriners Hospitals Pediatric Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States.,Department of Neurology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| |
Collapse
|
14
|
Nazri MUIA, Idris I, Ross O, Ismail WIW. Neurological Disorder Brain Model: A Lesson from Marine Worms (Annelida: Polychaeta). Malays J Med Sci 2019; 26:5-18. [PMID: 31908583 PMCID: PMC6939724 DOI: 10.21315/mjms2019.26.6.2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Accepted: 07/09/2019] [Indexed: 12/25/2022] Open
Abstract
The incidence of neurodegenerative diseases is directly proportional to age. The prevalence of non-communicable diseases, for example, Alzheimer's and Parkinson's, is expected to rise in the coming years. Understanding the etiopathology of these diseases is a crucial step that needs to be taken to develop drugs for their treatment. Animal models are being increasingly used to expand the knowledge and understanding on neurodegenerative diseases. Marine worms, known as polychaetes (phylum Annelida), which are abundantly and frequently found in benthic environments, possess a simple yet complete nervous system (including a true brain that is centralised and specialised) compared to other annelids. Hence, polychaetes can potentially be the next candidate for a nerve disease model. The ability to activate the entire nervous system regeneration (NSR) is among the remarkable features of many polychaetes species. However, the information on NSR in polychaetes and how it can potentially model neurodegenerative diseases in humans is still lacking. By exploring such studies, we may eventually be able to circumvent the developmental constraints that limit NSR in the human nervous system. This article is intended to briefly review responsible mechanisms and signalling pathways of NSR in marine polychaetes and to make a comparison with other established models of neurodegenerative disease.
Collapse
Affiliation(s)
| | - Izwandy Idris
- Institute of Oceanography and Environment, Universiti Malaysia Terengganu, Terengganu, Malaysia
| | - Othman Ross
- Institute of Oceanography and Environment, Universiti Malaysia Terengganu, Terengganu, Malaysia
| | - Wan Iryani Wan Ismail
- Faculty of Science and Marine Environment, Universiti Malaysia Terengganu, Terengganu, Malaysia
| |
Collapse
|
15
|
Jakimovski D, Zivadinov R, Ramanthan M, Hagemeier J, Weinstock-Guttman B, Tomic D, Kropshofer H, Fuchs TA, Barro C, Leppert D, Yaldizli Ö, Kuhle J, Benedict RHB. Serum neurofilament light chain level associations with clinical and cognitive performance in multiple sclerosis: A longitudinal retrospective 5-year study. Mult Scler 2019; 26:1670-1681. [DOI: 10.1177/1352458519881428] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Background: A limited number of studies investigated associations between serum neurofilament light chain (sNfL) and cognition in persons with multiple sclerosis (PwMS). Objective: To assess cross-sectional and longitudinal associations between sNfL levels, clinical, and cognitive performance in PwMS and age-matched healthy controls (HCs). Materials: One hundred twenty-seven PwMS (85 relapsing–remitting MS/42 progressive MS), 20 clinically isolated syndrome patients, and 52 HCs were followed for 5 years. sNfL levels were measured using the single-molecule array (Simoa) assay and quantified in picograms per milliliter. Expanded Disability Status Scale (EDSS), walking, and manual dexterity tests were obtained. At follow-up, Brief International Cognitive Assessment for MS (BICAMS) was utilized. Cognitively impaired (CI) status was derived using HC-based z-scores. Age-, sex-, and education-adjusted analysis of covariance (ANCOVA) and regression models were used. Multiple comparison–adjusted values of q < 0.05 were considered significant. Results: In PwMS, sNfL levels were cross-sectionally associated with walking speed ( r = 0.235, q = 0.036), manual dexterity ( r = 0.337, q = 0.002), and cognitive processing speed (CPS; r =−0.265, q = 0.012). Baseline sNfL levels predicted 5-year EDSS scores ( r = 0.25, q = 0.012), dexterity ( r = 0.224, q = 0.033), and CPS ( r =−0.205, q = 0.049). CI patients had higher sNfL levels (27.2 vs. 20.6, p = 0.016) and greater absolute longitudinal sNfL increase when compared with non-CI patients (4.8 vs. 0.7, p = 0.04). Conclusion: Higher sNfL levels are associated with poorer current and future clinical and cognitive performance.
Collapse
Affiliation(s)
- Dejan Jakimovski
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA/Jacobs MS Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Robert Zivadinov
- Jacobs MS Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA/Center for Biomedical Imaging at Clinical Translational Science Institute, Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Murali Ramanthan
- Department of Pharmaceutical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Jesper Hagemeier
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Bianca Weinstock-Guttman
- Jacobs MS Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | | | | | - Tom A Fuchs
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Christian Barro
- Neurologic Clinic and Policlinic, Departments of Medicine, Biomedicine, and Clinical Research, University Hospital Basel, University of Basel, Basel, Switzerland
| | | | - Özgür Yaldizli
- Neurologic Clinic and Policlinic, Departments of Medicine, Biomedicine, and Clinical Research, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Jens Kuhle
- Neurologic Clinic and Policlinic, Departments of Medicine, Biomedicine, and Clinical Research, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Ralph HB Benedict
- Jacobs MS Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| |
Collapse
|
16
|
MiR-105 and miR-9 regulate the mRNA stability of neuronal intermediate filaments. Implications for the pathogenesis of amyotrophic lateral sclerosis (ALS). Brain Res 2019; 1706:93-100. [DOI: 10.1016/j.brainres.2018.10.032] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 10/19/2018] [Accepted: 10/28/2018] [Indexed: 12/19/2022]
|
17
|
Morgan S, Duguez S, Duddy W. Personalized Medicine and Molecular Interaction Networks in Amyotrophic Lateral Sclerosis (ALS): Current Knowledge. J Pers Med 2018; 8:E44. [PMID: 30551677 PMCID: PMC6313785 DOI: 10.3390/jpm8040044] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 11/27/2018] [Accepted: 12/05/2018] [Indexed: 12/12/2022] Open
Abstract
Multiple genes and mechanisms of pathophysiology have been implicated in amyotrophic lateral sclerosis (ALS), suggesting it is a complex systemic disease. With this in mind, applying personalized medicine (PM) approaches to tailor treatment pipelines for ALS patients may be necessary. The modelling and analysis of molecular interaction networks could represent valuable resources in defining ALS-associated pathways and discovering novel therapeutic targets. Here we review existing omics datasets and analytical approaches, in order to consider how molecular interaction networks could improve our understanding of the molecular pathophysiology of this fatal neuromuscular disorder.
Collapse
Affiliation(s)
- Stephen Morgan
- Northern Ireland Centre for Stratified Medicine, Altnagelvin Hospital Campus, Ulster University, Londonderry, BT47 6SB, Northern Ireland, UK.
| | - Stephanie Duguez
- Northern Ireland Centre for Stratified Medicine, Altnagelvin Hospital Campus, Ulster University, Londonderry, BT47 6SB, Northern Ireland, UK.
| | - William Duddy
- Northern Ireland Centre for Stratified Medicine, Altnagelvin Hospital Campus, Ulster University, Londonderry, BT47 6SB, Northern Ireland, UK.
| |
Collapse
|
18
|
Khalil M, Teunissen CE, Otto M, Piehl F, Sormani MP, Gattringer T, Barro C, Kappos L, Comabella M, Fazekas F, Petzold A, Blennow K, Zetterberg H, Kuhle J. Neurofilaments as biomarkers in neurological disorders. Nat Rev Neurol 2018; 14:577-589. [DOI: 10.1038/s41582-018-0058-z] [Citation(s) in RCA: 767] [Impact Index Per Article: 109.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
19
|
Campos-Melo D, Hawley ZCE, Strong MJ. Dysregulation of human NEFM and NEFH mRNA stability by ALS-linked miRNAs. Mol Brain 2018; 11:43. [PMID: 30029677 PMCID: PMC6054723 DOI: 10.1186/s13041-018-0386-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 07/03/2018] [Indexed: 12/12/2022] Open
Abstract
Neurofilaments (NFs) are the most abundant cytoskeletal component of vertebrate myelinated axons. NFs function by determining axonal caliber, promoting axonal growth and forming a 3-dimensional lattice that supports the organization of cytoplasmic organelles. The stoichiometry of NF protein subunits (NFL, NFM and NFH) has to be tightly controlled to avoid the formation of NF neuronal cytoplasmic inclusions (NCIs), axonal degeneration and neuronal death, all pathological hallmarks of amyotrophic lateral sclerosis (ALS). The post-transcriptional control of NF transcripts is critical for regulating normal levels of NF proteins. Previously, we showed that miRNAs that are dysregulated in ALS spinal cord regulate the levels of NEFL mRNA. In order to complete the understanding of altered NF expression in ALS, in this study we have investigated the regulation of NEFM and NEFH mRNA levels by miRNAs. We observed that a small group of ALS-linked miRNAs that are expressed in human spinal motor neurons directly regulate NEFM and NEFH transcript levels in a manner that is associated with an increase in NFM and NFH protein levels in ALS spinal cord homogenates. In concert with previous observations demonstrating the suppression of NEFL mRNA steady state levels in ALS, these observations provide support for the hypothesis that the dysregulation of miRNAs in spinal motor neurons in ALS fundamentally alters the stoichiometry of NF expression, leading to the formation of pathological NCIs.
Collapse
Affiliation(s)
- Danae Campos-Melo
- Molecular Medicine Group, Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Zachary C E Hawley
- Molecular Medicine Group, Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Michael J Strong
- Molecular Medicine Group, Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada. .,Department of Pathology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada. .,Department of Clinical Neurological Sciences, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada. .,University Hospital, LHSC, Rm C7-120, 339, Windermere Road, London, ON, N6A 5A5, Canada.
| |
Collapse
|
20
|
Adiutori R, Aarum J, Zubiri I, Bremang M, Jung S, Sheer D, Pike I, Malaspina A. The proteome of neurofilament-containing protein aggregates in blood. Biochem Biophys Rep 2018; 14:168-177. [PMID: 29872749 PMCID: PMC5986704 DOI: 10.1016/j.bbrep.2018.04.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 03/15/2018] [Accepted: 04/26/2018] [Indexed: 11/24/2022] Open
Abstract
Protein aggregation in biofluids is a poorly understood phenomenon. Under normal physiological conditions, fluid-borne aggregates may contain plasma or cell proteins prone to aggregation. Recent observations suggest that neurofilaments (Nf), the building blocks of neurons and a biomarker of neurodegeneration, are included in high molecular weight complexes in circulation. The composition of these Nf-containing hetero-aggregates (NCH) may change in systemic or organ-specific pathologies, providing the basis to develop novel disease biomarkers. We have tested ultracentrifugation (UC) and a commercially available protein aggregate binder, Seprion PAD-Beads (SEP), for the enrichment of NCH from plasma of healthy individuals, and then characterised the Nf content of the aggregate fractions using gel electrophoresis and their proteome by mass spectrometry (MS). Western blot analysis of fractions obtained by UC showed that among Nf isoforms, neurofilament heavy chain (NfH) was found within SDS-stable high molecular weight aggregates. Shotgun proteomics of aggregates obtained with both extraction techniques identified mostly cell structural and to a lesser extent extra-cellular matrix proteins, while functional analysis revealed pathways involved in inflammatory response, phagosome and prion-like protein behaviour. UC aggregates were specifically enriched with proteins involved in endocrine, metabolic and cell-signalling regulation. We describe the proteome of neurofilament-containing aggregates isolated from healthy individuals biofluids using different extraction methods.
Collapse
Affiliation(s)
- Rocco Adiutori
- Centre for Neuroscience and Trauma, Queen Mary University of London, Blizard Institute, Barts and The School of Medicine and Dentistry, London, United Kingdom
| | - Johan Aarum
- Centre for Genomics and Child Health, Queen Mary University of London, Blizard Institute, Barts and The London School of Medicine and Dentistry, London, United Kingdom
| | - Irene Zubiri
- Centre for Neuroscience and Trauma, Queen Mary University of London, Blizard Institute, Barts and The School of Medicine and Dentistry, London, United Kingdom
| | - Michael Bremang
- Proteome Sciences Plc, Hamilton House, Mabledon Place, London, United Kingdom
| | - Stephan Jung
- ProteomeSciencesR&DGmbH&Co.KG, Frankfurt, Germany
| | - Denise Sheer
- Centre for Genomics and Child Health, Queen Mary University of London, Blizard Institute, Barts and The London School of Medicine and Dentistry, London, United Kingdom
| | - Ian Pike
- Proteome Sciences Plc, Hamilton House, Mabledon Place, London, United Kingdom
| | - Andrea Malaspina
- Centre for Neuroscience and Trauma, Queen Mary University of London, Blizard Institute, Barts and The School of Medicine and Dentistry, London, United Kingdom
| |
Collapse
|
21
|
Lu J, Lu L, Yu Y, Cluette-Brown J, Martin CR, Claud EC. Effects of Intestinal Microbiota on Brain Development in Humanized Gnotobiotic Mice. Sci Rep 2018; 8:5443. [PMID: 29615691 PMCID: PMC5882882 DOI: 10.1038/s41598-018-23692-w] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 03/19/2018] [Indexed: 12/23/2022] Open
Abstract
Poor growth in the Neonatal Intensive Care Unit is associated with an increased risk for poor neurodevelopmental outcomes for preterm infants, however the mechanism is unclear. The microbiome has increasingly been recognized as a modifiable environmental factor to influence host development. Here we explore the hypothesis that the microbiome influences both growth phenotype and brain development. A germ free mouse transfaunation model was used to examine the effects of preterm infant microbiotas known to induce either high growth or low growth phenotypes on postnatal brain development. The microbiome which induced the low growth phenotype was associated with decreases in the neuronal markers NeuN and neurofilament-L as well as the myelination marker MBP when compared to the microbiome associated with the high growth phenotype. Additionally, poor growth phenotype-associated microbiota was associated with increased neuroinflammation marked by increased Nos1, as well as alteration in IGF-1 pathway including decreased circulating and brain IGF-1, decreased circulating IGFBP3, and increased Igfbp3 brain mRNA expression. This study suggests that growth-associated microbiota can influence early neuron and oligodendrocyte development and that this effect may be mediated by effects on neuroinflammation and circulating IGF-1.
Collapse
Affiliation(s)
- Jing Lu
- The University of Chicago, Pritzker School of Medicine, Department of Pediatrics, Chicago, IL, 60637, USA
| | - Lei Lu
- The University of Chicago, Pritzker School of Medicine, Department of Pediatrics, Chicago, IL, 60637, USA
| | - Yueyue Yu
- The University of Chicago, Pritzker School of Medicine, Department of Pediatrics, Chicago, IL, 60637, USA
| | - Joanne Cluette-Brown
- Beth Israel Deaconess Medical Center, Division of Gastroenterology, Boston, MA, 02215, USA
| | - Camilia R Martin
- Beth Israel Deaconess Medical Center, Harvard Medical School, Department of Neonatology and Division of Translational Research, Boston, MA, 02215, USA
| | - Erika C Claud
- The University of Chicago, Pritzker School of Medicine, Department of Pediatrics, Chicago, IL, 60637, USA.
| |
Collapse
|
22
|
Fialova L, Bartos A, Svarcova J. Neurofilaments and tau proteins in cerebrospinal fluid and serum in dementias and neuroinflammation. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2017; 161:286-295. [DOI: 10.5507/bp.2017.038] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 09/04/2017] [Indexed: 12/12/2022] Open
|
23
|
Whitworth GB, Misaghi BC, Rosenthal DM, Mills EA, Heinen DJ, Watson AH, Ives CW, Ali SH, Bezold K, Marsh-Armstrong N, Watson FL. Translational profiling of retinal ganglion cell optic nerve regeneration in Xenopus laevis. Dev Biol 2017; 426:360-373. [PMID: 27471010 PMCID: PMC5897040 DOI: 10.1016/j.ydbio.2016.06.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 06/01/2016] [Accepted: 06/01/2016] [Indexed: 11/29/2022]
Abstract
Unlike adult mammals, adult frogs regrow their optic nerve following a crush injury, making Xenopus laevis a compelling model for studying the molecular mechanisms that underlie neuronal regeneration. Using Translational Ribosome Affinity Purification (TRAP), a method to isolate ribosome-associated mRNAs from a target cell population, we have generated a transcriptional profile by RNA-Seq for retinal ganglion cells (RGC) during the period of recovery following an optic nerve injury. Based on bioinformatic analysis using the Xenopus laevis 9.1 genome assembly, our results reveal a profound shift in the composition of ribosome-associated mRNAs during the early stages of RGC regeneration. As factors involved in cell signaling are rapidly down-regulated, those involved in protein biosynthesis are up-regulated alongside key initiators of axon development. Using the new genome assembly, we were also able to analyze gene expression profiles of homeologous gene pairs arising from a whole-genome duplication in the Xenopus lineage. Here we see evidence of divergence in regulatory control among a significant proportion of pairs. Our data should provide a valuable resource for identifying genes involved in the regeneration process to target for future functional studies, in both naturally regenerative and non-regenerative vertebrates.
Collapse
Affiliation(s)
- G B Whitworth
- Department of Biology, Washington and Lee University, Lexington, VA, United States
| | - B C Misaghi
- Department of Biology, Washington and Lee University, Lexington, VA, United States
| | - D M Rosenthal
- Department of Biology, Washington and Lee University, Lexington, VA, United States
| | - E A Mills
- Johns Hopkins University School of Medicine, Solomon H. Snyder Dept. of Neuroscience and Hugo Moser Research Institute at Kennedy Krieger, Baltimore, MD, United States
| | - D J Heinen
- Department of Biology, Washington and Lee University, Lexington, VA, United States
| | - A H Watson
- Department of Biology, Washington and Lee University, Lexington, VA, United States
| | - C W Ives
- Department of Biology, Washington and Lee University, Lexington, VA, United States
| | - S H Ali
- Department of Biology, Washington and Lee University, Lexington, VA, United States
| | - K Bezold
- Department of Biology, Washington and Lee University, Lexington, VA, United States
| | - N Marsh-Armstrong
- Johns Hopkins University School of Medicine, Solomon H. Snyder Dept. of Neuroscience and Hugo Moser Research Institute at Kennedy Krieger, Baltimore, MD, United States
| | - F L Watson
- Department of Biology, Washington and Lee University, Lexington, VA, United States.
| |
Collapse
|
24
|
Kirkcaldie MTK, Dwyer ST. The third wave: Intermediate filaments in the maturing nervous system. Mol Cell Neurosci 2017; 84:68-76. [PMID: 28554564 DOI: 10.1016/j.mcn.2017.05.010] [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: 02/20/2017] [Revised: 05/10/2017] [Accepted: 05/25/2017] [Indexed: 01/16/2023] Open
Abstract
Intermediate filaments are critical for the extreme structural specialisations of neurons, providing integrity in dynamic environments and efficient communication along axons a metre or more in length. As neurons mature, an initial expression of nestin and vimentin gives way to the neurofilament triplet proteins and α-internexin, substituted by peripherin in axons outside the CNS, which physically consolidate axons as they elongate and find their targets. Once connection is established, these proteins are transported, assembled, stabilised and modified, structurally transforming axons and dendrites as they acquire their full function. The interaction between these neurons and myelinating glial cells optimises the structure of axons for peak functional efficiency, a property retained across their lifespan. This finely calibrated structural regulation allows the nervous system to maintain timing precision and efficient control across large distances throughout somatic growth and, in maturity, as a plasticity mechanism allowing functional adaptation.
Collapse
Affiliation(s)
- Matthew T K Kirkcaldie
- School of Medicine, Wicking Dementia Research and Education Centre, Faculty of Health, University of Tasmania, Australia.
| | - Samuel T Dwyer
- School of Medicine, Wicking Dementia Research and Education Centre, Faculty of Health, University of Tasmania, Australia
| |
Collapse
|
25
|
Hawley ZCE, Campos-Melo D, Droppelmann CA, Strong MJ. MotomiRs: miRNAs in Motor Neuron Function and Disease. Front Mol Neurosci 2017; 10:127. [PMID: 28522960 PMCID: PMC5415563 DOI: 10.3389/fnmol.2017.00127] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Accepted: 04/18/2017] [Indexed: 12/12/2022] Open
Abstract
MiRNAs are key regulators of the mammalian transcriptome that have been increasingly linked to degenerative diseases of the motor neurons. Although many of the miRNAs currently incriminated as participants in the pathogenesis of these diseases are also important to the normal development and function of motor neurons, at present there is no knowledge of the complete miRNA profile of motor neurons. In this review, we examine the current understanding with respect to miRNAs that are specifically required for motor neuron development, function and viability, and provide evidence that these should be considered as a functional network of miRNAs which we have collectively termed MotomiRs. We will also summarize those MotomiRs currently known to be associated with both amyotrophic lateral sclerosis (ALS) and spinal muscular atrophy (SMA), and discuss their potential use as biomarkers.
Collapse
Affiliation(s)
- Zachary C E Hawley
- Molecular Medicine Group, Robarts Research Institute, Schulich School of Medicine and Dentistry, Western UniversityLondon, ON, Canada
| | - Danae Campos-Melo
- Molecular Medicine Group, Robarts Research Institute, Schulich School of Medicine and Dentistry, Western UniversityLondon, ON, Canada
| | - Cristian A Droppelmann
- Molecular Medicine Group, Robarts Research Institute, Schulich School of Medicine and Dentistry, Western UniversityLondon, ON, Canada
| | - Michael J Strong
- Molecular Medicine Group, Robarts Research Institute, Schulich School of Medicine and Dentistry, Western UniversityLondon, ON, Canada.,Department of Pathology, Schulich School of Medicine and Dentistry, Western UniversityLondon, ON, Canada.,Department of Clinical Neurological Sciences, Schulich School of Medicine and Dentistry, Western UniversityLondon, ON, Canada
| |
Collapse
|
26
|
Rosso G, Liashkovich I, Young P, Röhr D, Shahin V. Schwann cells and neurite outgrowth from embryonic dorsal root ganglions are highly mechanosensitive. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2017; 13:493-501. [DOI: 10.1016/j.nano.2016.06.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Revised: 06/06/2016] [Accepted: 06/24/2016] [Indexed: 02/02/2023]
|
27
|
Hutchins EJ, Belrose JL, Szaro BG. A novel role for the nuclear localization signal in regulating hnRNP K protein stability in vivo. Biochem Biophys Res Commun 2016; 478:772-6. [DOI: 10.1016/j.bbrc.2016.08.023] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 08/04/2016] [Indexed: 01/12/2023]
|
28
|
Neurofilament depletion improves microtubule dynamics via modulation of Stat3/stathmin signaling. Acta Neuropathol 2016; 132:93-110. [PMID: 27021905 PMCID: PMC4911381 DOI: 10.1007/s00401-016-1564-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 03/14/2016] [Accepted: 03/15/2016] [Indexed: 12/31/2022]
Abstract
In neurons, microtubules form a dense array within axons, and the stability and function of this microtubule network is modulated by neurofilaments. Accumulation of neurofilaments has been observed in several forms of neurodegenerative diseases, but the mechanisms how elevated neurofilament levels destabilize axons are unknown so far. Here, we show that increased neurofilament expression in motor nerves of pmn mutant mice, a model of motoneuron disease, causes disturbed microtubule dynamics. The disease is caused by a point mutation in the tubulin-specific chaperone E (Tbce) gene, leading to an exchange of the most C-terminal amino acid tryptophan to glycine. As a consequence, the TBCE protein becomes instable which then results in destabilization of axonal microtubules and defects in axonal transport, in particular in motoneurons. Depletion of neurofilament increases the number and regrowth of microtubules in pmn mutant motoneurons and restores axon elongation. This effect is mediated by interaction of neurofilament with the stathmin complex. Accumulating neurofilaments associate with stathmin in axons of pmn mutant motoneurons. Depletion of neurofilament by Nefl knockout increases Stat3–stathmin interaction and stabilizes the microtubules in pmn mutant motoneurons. Consequently, counteracting enhanced neurofilament expression improves axonal maintenance and prolongs survival of pmn mutant mice. We propose that this mechanism could also be relevant for other neurodegenerative diseases in which neurofilament accumulation and loss of microtubules are prominent features.
Collapse
|
29
|
Increase of neurofilament-H protein in sensory neurons in antiretroviral neuropathy: Evidence for a neuroprotective response mediated by the RNA-binding protein HuD. Pharmacol Res 2016; 111:23-33. [PMID: 27238228 DOI: 10.1016/j.phrs.2016.05.026] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 05/06/2016] [Accepted: 05/25/2016] [Indexed: 01/31/2023]
Abstract
Nucleoside reverse transcriptase inhibitors (NRTIs) are key components of HIV/AIDS treatment to reduce viral load. However, antiretroviral toxic neuropathy has become a common peripheral neuropathy among HIV/AIDS patients leading to discontinuation of antiretroviral therapy, for which the underlying pathogenesis is uncertain. This study examines the role of neurofilament (NF) proteins in the spinal dorsal horn, DRG and sciatic nerve after NRTI neurotoxicity in mice treated with zalcitabine (2',3'-dideoxycitidine; ddC). ddC administration up-regulated NF-M and pNF-H proteins with no effect on NF-L. The increase of pNF-H levels was counteracted by the silencing of HuD, an RNA binding protein involved in neuronal development and differentiation. Sciatic nerve sections of ddC exposed mice showed an increased axonal caliber, concomitantly to a pNF-H up-regulation. Both events were prevented by HuD silencing. pNF-H and HuD colocalize in DRG and spinal dorsal horn axons. However, the capability of HuD to bind NF mRNA was not demonstrated, indicating the presence of an indirect mechanism of control of NF expression by HuD. RNA immunoprecipitation experiments showed the capability of HuD to bind the BDNF mRNA and the administration of an anti-BDNF antibody prevented pNF-H increase. These data indicate the presence of a HuD - BDNF - NF-H pathway activated as a regenerative response to the axonal damage induced by ddC treatment to counteract the antiretroviral neurotoxicity. Since analgesics clinically used to treat neuropathic pain are ineffective on antiretroviral neuropathy, a neuroregenerative strategy might represent a new therapeutic opportunity to counteract neurotoxicity and avoid discontinuation or abandon of NRTI therapy.
Collapse
|
30
|
Wang C, Szaro BG. Post-transcriptional regulation mediated by specific neurofilament introns in vivo. J Cell Sci 2016; 129:1500-11. [PMID: 26906423 DOI: 10.1242/jcs.185199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 02/17/2016] [Indexed: 11/20/2022] Open
Abstract
Neurons regulate genes post-transcriptionally to coordinate the supply of cytoskeletal proteins, such as the medium neurofilament (NEFM), with demand for structural materials in response to extracellular cues encountered by developing axons. By using a method for evaluating functionality of cis-regulatory gene elements in vivo through plasmid injection into Xenopus embryos, we discovered that splicing of a specific nefm intron was required for robust transgene expression, regardless of promoter or cell type. Transgenes utilizing the nefm 3'-UTR but substituting other nefm introns expressed little or no protein owing to defects in handling of the messenger (m)RNA as opposed to transcription or splicing. Post-transcriptional events at multiple steps, but mainly during nucleocytoplasmic export, contributed to these varied levels of protein expression. An intron of the β-globin gene was also able to promote expression in a manner identical to that of the nefm intron, implying a more general preference for certain introns in controlling nefm expression. These results expand our knowledge of intron-mediated gene expression to encompass neurofilaments, indicating an additional layer of complexity in the control of a cytoskeletal gene needed for developing and maintaining healthy axons.
Collapse
Affiliation(s)
- Chen Wang
- Department of Biological Sciences and the Center for Neuroscience Research, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY 12222, USA
| | - Ben G Szaro
- Department of Biological Sciences and the Center for Neuroscience Research, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY 12222, USA
| |
Collapse
|
31
|
Sassone J, Taiana M, Lombardi R, Porretta-Serapiglia C, Freschi M, Bonanno S, Marcuzzo S, Caravello F, Bendotti C, Lauria G. ALS mouse model SOD1G93A displays early pathology of sensory small fibers associated to accumulation of a neurotoxic splice variant of peripherin. Hum Mol Genet 2016; 25:1588-99. [PMID: 26908600 DOI: 10.1093/hmg/ddw035] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2015] [Accepted: 02/05/2016] [Indexed: 12/12/2022] Open
Abstract
Growing evidence suggests that amyotrophic lateral sclerosis (ALS) is a multisystem neurodegenerative disease that primarily affects motor neurons and, though less evidently, other neuronal systems. About 75% of sporadic and familial ALS patients show a subclinical degeneration of small-diameter fibers, as measured by loss of intraepidermal nerve fibers (IENFs), but the underlying biological causes are unknown. Small-diameter fibers are derived from small-diameter sensory neurons, located in dorsal root ganglia (DRG), whose biochemical hallmark is the expression of type III intermediate filament peripherin. We tested here the hypothesis that small-diameter DRG neurons of ALS mouse model SOD1(G93A)suffer from axonal stress and investigated the underlying molecular mechanism. We found that SOD1(G93A)mice display small fiber pathology, as measured by IENF loss, which precedes the onset of the disease. In vitro small-diameter DRG neurons of SOD1(G93A)mice show axonal stress features and accumulation of a peripherin splice variant, named peripherin56, which causes axonal stress through disassembling light and medium neurofilament subunits (NFL and NFM, respectively). Our findings first demonstrate that small-diameter DRG neurons of the ALS mouse model SOD1(G93A)display axonal stress in vitro and in vivo, thus sustaining the hypothesis that the effects of ALS disease spread beyond motor neurons. These results suggest a molecular mechanism for the small fiber pathology found in ALS patients. Finally, our data agree with previous findings, suggesting a key role of peripherin in the ALS pathogenesis, thus highlighting that DRG neurons mirror some dysfunctions found in motor neurons.
Collapse
Affiliation(s)
| | | | | | | | - Mattia Freschi
- Neuroscience Department, Laboratory of Molecular Neurobiology, IRCCS Istituto di Ricerche Farmacologiche 'Mario Negri', Animal Facility Fondazione italiana per la ricerca sulla SLA (AriSLA), Milan, Italy and
| | - Silvia Bonanno
- 4th Neurology Unit, IRCCS Foundation 'Carlo Besta' Neurological Institute, via Celoria 11, 20133 Milan, Italy, PhD Program in Neuroscience, University of Milan, Bicocca, Italy
| | - Stefania Marcuzzo
- 4th Neurology Unit, IRCCS Foundation 'Carlo Besta' Neurological Institute, via Celoria 11, 20133 Milan, Italy
| | | | - Caterina Bendotti
- Neuroscience Department, Laboratory of Molecular Neurobiology, IRCCS Istituto di Ricerche Farmacologiche 'Mario Negri'
| | | |
Collapse
|
32
|
Ferri E, Arosio B, D'Addario C, Galimberti D, Gussago C, Pucci M, Casati M, Fenoglio C, Abbate C, Rossi PD, Scarpini E, Maccarrone M, Mari D. Gene promoter methylation and expression of Pin1 differ between patients with frontotemporal dementia and Alzheimer's disease. J Neurol Sci 2016; 362:283-6. [PMID: 26944164 DOI: 10.1016/j.jns.2016.02.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 01/07/2016] [Accepted: 02/02/2016] [Indexed: 11/17/2022]
Abstract
Frontotemporal Dementia (FTD) and Alzheimer's Disease (AD) share the accumulation of fibrillar aggregates of misfolded proteins. To better understand these neurodegenerative diseases and identify biomarkers in easily accessible cells, we investigated DNA methylation at Pin1 gene promoter and its expression in peripheral blood mononuclear cells of FTD patients. We found a lower gene expression of Pin1 with a higher DNA methylation in three CpG sites at Pin1 gene promoter analysed in FTD subjects, in contrast to a higher gene expression with a lower methylation in AD subjects and controls. These data suggest an important and distinct involvement of Pin1 in these two types of dementia.
Collapse
Affiliation(s)
- Evelyn Ferri
- Geriatric Unit, Department of Medical Sciences and Community Health, University of Milan, Via Pace 9, 20122 Milan, Italy; PhD in Nutritional Sciences University of Milan, Via Festa del Perdono 7, 20122 Milan, Italy.
| | - Beatrice Arosio
- Geriatric Unit, Department of Medical Sciences and Community Health, University of Milan, Via Pace 9, 20122 Milan, Italy; Fondazione Ca' Granda, IRCCS Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122 Milan, Italy
| | - Claudio D'Addario
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Piazza Aldo Moro 45, 64100 Teramo, Italy; European Center for Brain Research (CERC)/Santa Lucia Foundation, Via del Fosso di Fiorano 64, 00143 Rome, Italy
| | - Daniela Galimberti
- Fondazione Ca' Granda, IRCCS Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122 Milan, Italy; Department of Pathophysiology and Transplantation, "Dino Ferrari" Center, University of Milan, Via Francesco Sforza 35, 20122 Milan, Italy
| | - Cristina Gussago
- Geriatric Unit, Department of Medical Sciences and Community Health, University of Milan, Via Pace 9, 20122 Milan, Italy
| | - Mariangela Pucci
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Piazza Aldo Moro 45, 64100 Teramo, Italy
| | - Martina Casati
- Geriatric Unit, Department of Medical Sciences and Community Health, University of Milan, Via Pace 9, 20122 Milan, Italy; PhD in Nutritional Sciences University of Milan, Via Festa del Perdono 7, 20122 Milan, Italy
| | - Chiara Fenoglio
- Fondazione Ca' Granda, IRCCS Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122 Milan, Italy; Department of Pathophysiology and Transplantation, "Dino Ferrari" Center, University of Milan, Via Francesco Sforza 35, 20122 Milan, Italy
| | - Carlo Abbate
- Fondazione Ca' Granda, IRCCS Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122 Milan, Italy
| | - Paolo Dionigi Rossi
- Fondazione Ca' Granda, IRCCS Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122 Milan, Italy
| | - Elio Scarpini
- Fondazione Ca' Granda, IRCCS Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122 Milan, Italy; Department of Pathophysiology and Transplantation, "Dino Ferrari" Center, University of Milan, Via Francesco Sforza 35, 20122 Milan, Italy
| | - Mauro Maccarrone
- European Center for Brain Research (CERC)/Santa Lucia Foundation, Via del Fosso di Fiorano 64, 00143 Rome, Italy; Campus Bio-Medico University of Rome, Via Álvaro del Portillo 21, 00128 Rome, Italy
| | - Daniela Mari
- Geriatric Unit, Department of Medical Sciences and Community Health, University of Milan, Via Pace 9, 20122 Milan, Italy; Fondazione Ca' Granda, IRCCS Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122 Milan, Italy
| |
Collapse
|
33
|
Using Xenopus Embryos to Study Transcriptional and Posttranscriptional Gene Regulatory Mechanisms of Intermediate Filaments. Methods Enzymol 2016; 568:635-60. [DOI: 10.1016/bs.mie.2015.07.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023]
|
34
|
Reis KP, Heimfarth L, Pierozan P, Ferreira F, Loureiro SO, Fernandes CG, Carvalho RV, Pessoa-Pureur R. High postnatal susceptibility of hippocampal cytoskeleton in response to ethanol exposure during pregnancy and lactation. Alcohol 2015; 49:665-74. [PMID: 26314629 DOI: 10.1016/j.alcohol.2015.06.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Revised: 06/08/2015] [Accepted: 06/08/2015] [Indexed: 12/30/2022]
Abstract
Ethanol exposure to offspring during pregnancy and lactation leads to developmental disorders, including central nervous system dysfunction. In the present work, we have studied the effect of chronic ethanol exposure during pregnancy and lactation on the phosphorylating system associated with the astrocytic and neuronal intermediate filament (IF) proteins: glial fibrillary acidic protein (GFAP), and neurofilament (NF) subunits of low, medium, and high molecular weight (NFL, NFM, and NFH, respectively) in 9- and 21-day-old pups. Female rats were fed with 20% ethanol in their drinking water during pregnancy and lactation. The homeostasis of the IF phosphorylation was not altered in the cerebral cortex, cerebellum, or hippocampus of 9-day-old pups. However, GFAP, NFL, and NFM were hyperphosphorylated in the hippocampus of 21-day-old pups. PKA had been activated in the hippocampus, and Ser55 in the N-terminal region of NFL was hyperphosphorylated. In addition, JNK/MAPK was activated and KSP repeats in the C-terminal region of NFM were hyperphosphorylated in the hippocampus of 21-day-old pups. Decreased NFH immunocontent but an unaltered total NFH/phosphoNFH ratio suggested altered stoichiometry of NFs in the hippocampus of ethanol-exposed 21-day-old pups. In contrast to the high susceptibility of hippocampal cytoskeleton in developing rats, the homeostasis of the cytoskeleton of ethanol-fed adult females was not altered. Disruption of the cytoskeletal homeostasis in neural cells supports the view that regions of the brain are differentially vulnerable to alcohol insult during pregnancy and lactation, suggesting that modulation of JNK/MAPK and PKA signaling cascades target the hippocampal cytoskeleton in a window of vulnerability in 21-day-old pups. Our findings are relevant, since disruption of the cytoskeleton in immature hippocampus could contribute to later hippocampal damage associated with ethanol toxicity.
Collapse
Affiliation(s)
- Karina Pires Reis
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, Brazil
| | - Luana Heimfarth
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, Brazil
| | - Paula Pierozan
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, Brazil
| | - Fernanda Ferreira
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, Brazil
| | | | | | - Rônan Vivian Carvalho
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, Brazil
| | - Regina Pessoa-Pureur
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, Brazil.
| |
Collapse
|
35
|
Neurofilament light gene deletion exacerbates amyloid, dystrophic neurite, and synaptic pathology in the APP/PS1 transgenic model of Alzheimer's disease. Neurobiol Aging 2015; 36:2757-67. [DOI: 10.1016/j.neurobiolaging.2015.07.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 07/02/2015] [Accepted: 07/02/2015] [Indexed: 01/10/2023]
|
36
|
Piao CD, Yang K, Li P, Luo M. Autologous nerve graft repair of different degrees of sciatic nerve defect: stress and displacement at the anastomosis in a three-dimensional fnite element simulation model. Neural Regen Res 2015; 10:804-7. [PMID: 26109958 PMCID: PMC4468775 DOI: 10.4103/1673-5374.156986] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/16/2015] [Indexed: 02/06/2023] Open
Abstract
In the repair of peripheral nerve injury using autologous or synthetic nerve grafting, the magnitude of tensile forces at the anastomosis affects its response to physiological stress and the ultimate success of the treatment. One-dimensional stretching is commonly used to measure changes in tensile stress and strain; however, the accuracy of this simple method is limited. Therefore, in the present study, we established three-dimensional finite element models of sciatic nerve defects repaired by autologous nerve grafts. Using PRO E 5.0 finite element simulation software, we calculated the maximum stress and displacement of an anastomosis under a 5 N load in 10-, 20-, 30-, 40-mm long autologous nerve grafts. We found that maximum displacement increased with graft length, consistent with specimen force. These findings indicate that three-dimensional finite element simulation is a feasible method for analyzing stress and displacement at the anastomosis after autologous nerve grafting.
Collapse
Affiliation(s)
- Cheng-Dong Piao
- Department of Orthopedics, Second Hospital, Jilin University, Changchun, Jilin Province, China
| | - Kun Yang
- Basic Department, Air Force Aviation University of Chinese PLA, Changchun, Jilin Province, China
| | - Peng Li
- Department of Engineering Mechanics, Nanling Campus, Jilin University, Changchun, Jilin Province, China
| | - Min Luo
- Department of Pain, China-Japan Union Hospital, Jilin University, Changchun, Jilin Province, China
| |
Collapse
|
37
|
McGuire JL, Gill AJ, Douglas SD, Kolson DL. Central and peripheral markers of neurodegeneration and monocyte activation in HIV-associated neurocognitive disorders. J Neurovirol 2015; 21:439-48. [PMID: 25776526 PMCID: PMC4511078 DOI: 10.1007/s13365-015-0333-3] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Revised: 02/02/2015] [Accepted: 02/24/2015] [Indexed: 02/02/2023]
Abstract
HIV-associated neurocognitive disorders (HAND) affect up to 50 % of HIV-infected adults, independently predict HIV morbidity/mortality, and are associated with neuronal damage and monocyte activation. Cerebrospinal fluid (CSF) neurofilament subunits (NFL, pNFH) are sensitive surrogate markers of neuronal damage in several neurodegenerative diseases. In HIV, CSF NFL is elevated in individuals with and without cognitive impairment, suggesting early/persistent neuronal injury during HIV infection. Although individuals with severe cognitive impairment (HIV-associated dementia (HAD)) express higher CSF NFL levels than cognitively normal HIV-infected individuals, the relationships between severity of cognitive impairment, monocyte activation, neurofilament expression, and systemic infection are unclear. We performed a retrospective cross-sectional study of 48 HIV-infected adults with varying levels of cognitive impairment, not receiving antiretroviral therapy (ART), enrolled in the CNS Anti-Retroviral Therapy Effects Research (CHARTER) study. We quantified NFL, pNFH, and monocyte activation markers (sCD14/sCD163) in paired CSF/plasma samples. By examining subjects off ART, these correlations are not confounded by possible effects of ART on inflammation and neurodegeneration. We found that CSF NFL levels were elevated in individuals with HAD compared to cognitively normal or mildly impaired individuals with CD4+ T-lymphocyte nadirs ≤200. In addition, CSF NFL levels were significantly positively correlated to plasma HIV-1 RNA viral load and negatively correlated to plasma CD4+ T-lymphocyte count, suggesting a link between neuronal injury and systemic HIV infection. Finally, CSF NFL was significantly positively correlated with CSF pNFH, sCD163, and sCD14, demonstrating that monocyte activation within the CNS compartment is directly associated with neuronal injury at all stages of HAND.
Collapse
Affiliation(s)
- Jennifer L McGuire
- Division of Neurology, The Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA,
| | | | | | | |
Collapse
|
38
|
Laser-Azogui A, Kornreich M, Malka-Gibor E, Beck R. Neurofilament assembly and function during neuronal development. Curr Opin Cell Biol 2015; 32:92-101. [PMID: 25635910 DOI: 10.1016/j.ceb.2015.01.003] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 01/06/2015] [Accepted: 01/09/2015] [Indexed: 02/06/2023]
Abstract
Studies on the assembly of neuronal intermediate filaments (IFs) date back to the early work of Alzheimer. Developing neurons express a series of IF proteins, sequentially, at distinct stages of mammalian cell differentiation. This correlates with altered morphologies during the neuronal development, including axon outgrowth, guidance and conductivity. Importantly, neuronal IFs that fail to properly assemble into a filamentous network are a hallmark of neurodegenerative diseases such as amyotrophic lateral sclerosis, Alzheimer's, and Parkinson's disease. Traditional structural methodologies fail to fully describe neuronal IF assembly, interactions and resulting function due to IFs structural plasticity, particularly in their C-terminal domains. We review here current progress in the field of neuronal-specific IFs, a dominant component affecting the cytoskeletal structure and function of neurons.
Collapse
Affiliation(s)
- Adi Laser-Azogui
- The Raymond and Beverly Sackler School of Physics and Astronomy, Tel-Aviv University, Tel-Aviv 69978, Israel
| | - Micha Kornreich
- The Raymond and Beverly Sackler School of Physics and Astronomy, Tel-Aviv University, Tel-Aviv 69978, Israel
| | - Eti Malka-Gibor
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 69978, Israel
| | - Roy Beck
- The Raymond and Beverly Sackler School of Physics and Astronomy, Tel-Aviv University, Tel-Aviv 69978, Israel.
| |
Collapse
|
39
|
Droppelmann CA, Campos-Melo D, Volkening K, Strong MJ. The emerging role of guanine nucleotide exchange factors in ALS and other neurodegenerative diseases. Front Cell Neurosci 2014; 8:282. [PMID: 25309324 PMCID: PMC4159981 DOI: 10.3389/fncel.2014.00282] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 08/25/2014] [Indexed: 12/11/2022] Open
Abstract
Small GTPases participate in a broad range of cellular processes such as proliferation, differentiation, and migration. The exchange of GDP for GTP resulting in the activation of these GTPases is catalyzed by a group of enzymes called guanine nucleotide exchange factors (GEFs), of which two classes: Dbl-related exchange factors and the more recently described dedicator of cytokinesis proteins family exchange factors. Increasingly, deregulation of normal GEF activity or function has been associated with a broad range of disease states, including neurodegeneration and neurodevelopmental disorders. In this review, we examine this evidence with special emphasis on the novel role of Rho guanine nucleotide exchange factor (RGNEF/p190RhoGEF) in the pathogenesis of amyotrophic lateral sclerosis. RGNEF is the first neurodegeneration-linked GEF that regulates not only RhoA GTPase activation but also functions as an RNA binding protein that directly acts with low molecular weight neurofilament mRNA 3' untranslated region to regulate its stability. This dual role for RGNEF, coupled with the increasing understanding of the key role for GEFs in modulating the GTPase function in cell survival suggests a prominent role for GEFs in mediating a critical balance between cytotoxicity and neuroprotection which, when disturbed, contributes to neuronal loss.
Collapse
Affiliation(s)
- Cristian A Droppelmann
- Molecular Medicine Group, Robarts Research Institute, Western University London, ON, Canada
| | - Danae Campos-Melo
- Molecular Medicine Group, Robarts Research Institute, Western University London, ON, Canada
| | - Kathryn Volkening
- Molecular Medicine Group, Robarts Research Institute, Western University London, ON, Canada ; Department of Clinical Neurological Sciences, Schulich School of Medicine & Dentistry, Western University London, ON, Canada
| | - Michael J Strong
- Molecular Medicine Group, Robarts Research Institute, Western University London, ON, Canada ; Department of Clinical Neurological Sciences, Schulich School of Medicine & Dentistry, Western University London, ON, Canada
| |
Collapse
|
40
|
RNA-Binding Proteins Associated Molecular Mechanisms of Motor Neuron Degeneration Pathogenesis. Mol Biotechnol 2014; 56:779-86. [DOI: 10.1007/s12033-014-9785-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
41
|
Blizzard CA, King AE, Vickers J, Dickson T. Cortical murine neurons lacking the neurofilament light chain protein have an attenuated response to injury in vitro. J Neurotrauma 2014; 30:1908-18. [PMID: 23802559 DOI: 10.1089/neu.2013.2850] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Neurofilaments (NFs) have been proposed to have a significant role in attempted axonal regeneration following a variety of forms of injury. The NF triplet proteins of the central nervous system are comprised of light (NF-L), medium (NF-M) and heavy (NF-H) chains and are part of the type IV intermediate filament family. We sought to define the role of NF-L in the neuronal response to trauma and regeneration by examining the effect of total absence of the NF-L protein on neuronal maturation and response to axotomy. This study utilized an in vitro model comprising relatively mature cortical murine neurons derived from either wild-type embryonic (E15) mice or mice with a genetic knockout of NF-L (NF-L KO). Whilst NF-L KO neurons developed to relative maturity at a comparable rate to wild-type control neurons, NF-L KO neurons demonstrated relatively increased expression of α-internexin and decreased expression of NF-M. Further, we demonstrate that α-internexin co-immunoprecipitates with the NF binding protein NDel1 in NFL-KO cortical neurons in vitro. Following localized axotomy, NF-L KO neurons demonstrated reduced amyloid precursor protein accumulation in damaged neurites as well as a significant reduction in the number of axons regenerating (4.79+/-0.58 sprouts) in comparison to control preparations (10.47+/-1.11 sprouts) (p<0.05). These studies indicate that NFs comprising NF-L have a dynamic role in the reactive and regenerative changes in axons following injury.
Collapse
Affiliation(s)
- Catherine A Blizzard
- 1 Menzies Research Institute Tasmania, School of Medicine, University of Tasmania , Tasmania, Australia
| | | | | | | |
Collapse
|
42
|
Altered neuronal architecture and plasticity in the visual cortex of adult MMP-3-deficient mice. Brain Struct Funct 2014; 220:2675-89. [DOI: 10.1007/s00429-014-0819-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Accepted: 06/05/2014] [Indexed: 12/21/2022]
|
43
|
Wong TH, Chiu WZ, Breedveld GJ, Li KW, Verkerk AJMH, Hondius D, Hukema RK, Seelaar H, Frick P, Severijnen LA, Lammers GJ, Lebbink JHG, van Duinen SG, Kamphorst W, Rozemuller AJ, Bakker EB, Neumann M, Willemsen R, Bonifati V, Smit AB, van Swieten J. PRKAR1B mutation associated with a new neurodegenerative disorder with unique pathology. ACTA ACUST UNITED AC 2014; 137:1361-73. [PMID: 24722252 DOI: 10.1093/brain/awu067] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Pathological accumulation of intermediate filaments can be observed in neurodegenerative disorders, such as Alzheimer's disease, frontotemporal dementia and Parkinson's disease, and is also characteristic of neuronal intermediate filament inclusion disease. Intermediate filaments type IV include three neurofilament proteins (light, medium and heavy molecular weight neurofilament subunits) and α-internexin. The phosphorylation of intermediate filament proteins contributes to axonal growth, and is regulated by protein kinase A. Here we describe a family with a novel late-onset neurodegenerative disorder presenting with dementia and/or parkinsonism in 12 affected individuals. The disorder is characterized by a unique neuropathological phenotype displaying abundant neuronal inclusions by haematoxylin and eosin staining throughout the brain with immunoreactivity for intermediate filaments. Combining linkage analysis, exome sequencing and proteomics analysis, we identified a heterozygous c.149T>G (p.Leu50Arg) missense mutation in the gene encoding the protein kinase A type I-beta regulatory subunit (PRKAR1B). The pathogenicity of the mutation is supported by segregation in the family, absence in variant databases, and the specific accumulation of PRKAR1B in the inclusions in our cases associated with a specific biochemical pattern of PRKAR1B. Screening of PRKAR1B in 138 patients with Parkinson's disease and 56 patients with frontotemporal dementia did not identify additional novel pathogenic mutations. Our findings link a pathogenic PRKAR1B mutation to a novel hereditary neurodegenerative disorder and suggest an altered protein kinase A function through a reduced binding of the regulatory subunit to the A-kinase anchoring protein and the catalytic subunit of protein kinase A, which might result in subcellular dislocalization of the catalytic subunit and hyperphosphorylation of intermediate filaments.
Collapse
Affiliation(s)
- Tsz Hang Wong
- 1 Department of Neurology, Erasmus Medical Centre, 3015 CE Rotterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Chen H, Qian K, Du Z, Cao J, Petersen A, Liu H, Blackbourn LW, Huang CL, Errigo A, Yin Y, Lu J, Ayala M, Zhang SC. Modeling ALS with iPSCs reveals that mutant SOD1 misregulates neurofilament balance in motor neurons. Cell Stem Cell 2014; 14:796-809. [PMID: 24704493 DOI: 10.1016/j.stem.2014.02.004] [Citation(s) in RCA: 234] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2014] [Revised: 02/12/2014] [Accepted: 02/13/2014] [Indexed: 01/12/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) presents motoneuron (MN)-selective protein inclusions and axonal degeneration but the underlying mechanisms of such are unknown. Using induced pluripotent cells (iPSCs) from patients with mutation in the Cu/Zn superoxide dismutase (SOD1) gene, we show that spinal MNs, but rarely non-MNs, exhibited neurofilament (NF) aggregation followed by neurite degeneration when glia were not present. These changes were associated with decreased stability of NF-L mRNA and binding of its 3' UTR by mutant SOD1 and thus altered protein proportion of NF subunits. Such MN-selective changes were mimicked by expression of a single copy of the mutant SOD1 in human embryonic stem cells and were prevented by genetic correction of the SOD1 mutation in patient's iPSCs. Importantly, conditional expression of NF-L in the SOD1 iPSC-derived MNs corrected the NF subunit proportion, mitigating NF aggregation and neurite degeneration. Thus, NF misregulation underlies mutant SOD1-mediated NF aggregation and axonal degeneration in ALS MNs.
Collapse
Affiliation(s)
- Hong Chen
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China; Waisman Center, University of Wisconsin, Madison, WI 53705, USA
| | - Kun Qian
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China; Waisman Center, University of Wisconsin, Madison, WI 53705, USA
| | - Zhongwei Du
- Waisman Center, University of Wisconsin, Madison, WI 53705, USA
| | - Jingyuan Cao
- Waisman Center, University of Wisconsin, Madison, WI 53705, USA
| | - Andrew Petersen
- Waisman Center, University of Wisconsin, Madison, WI 53705, USA
| | - Huisheng Liu
- Waisman Center, University of Wisconsin, Madison, WI 53705, USA
| | | | | | - Anthony Errigo
- Waisman Center, University of Wisconsin, Madison, WI 53705, USA
| | - Yingnan Yin
- Waisman Center, University of Wisconsin, Madison, WI 53705, USA
| | - Jianfeng Lu
- Waisman Center, University of Wisconsin, Madison, WI 53705, USA
| | - Melvin Ayala
- Waisman Center, University of Wisconsin, Madison, WI 53705, USA
| | - Su-Chun Zhang
- Waisman Center, University of Wisconsin, Madison, WI 53705, USA; Department of Neuroscience and Department of Neurology, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53705, USA.
| |
Collapse
|
45
|
Fujita H, Aoki H, Ajioka I, Yamazaki M, Abe M, Oh-Nishi A, Sakimura K, Sugihara I. Detailed expression pattern of aldolase C (Aldoc) in the cerebellum, retina and other areas of the CNS studied in Aldoc-Venus knock-in mice. PLoS One 2014; 9:e86679. [PMID: 24475166 PMCID: PMC3903578 DOI: 10.1371/journal.pone.0086679] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Accepted: 12/13/2013] [Indexed: 11/22/2022] Open
Abstract
Aldolase C (Aldoc, also known as "zebrin II"), a brain type isozyme of a glycolysis enzyme, is expressed heterogeneously in subpopulations of cerebellar Purkinje cells (PCs) that are arranged longitudinally in a complex striped pattern in the cerebellar cortex, a pattern which is closely related to the topography of input and output axonal projections. Here, we generated knock-in Aldoc-Venus mice in which Aldoc expression is visualized by expression of a fluorescent protein, Venus. Since there was no obvious phenotypes in general brain morphology and in the striped pattern of the cerebellum in mutants, we made detailed observation of Aldoc expression pattern in the nervous system by using Venus expression in Aldoc-Venus heterozygotes. High levels of Venus expression were observed in cerebellar PCs, cartwheel cells in the dorsal cochlear nucleus, sensory epithelium of the inner ear and in all major types of retinal cells, while moderate levels of Venus expression were observed in astrocytes and satellite cells in the dorsal root ganglion. The striped arrangement of PCs that express Venus to different degrees was carefully traced with serial section alignment analysis and mapped on the unfolded scheme of the entire cerebellar cortex to re-identify all individual Aldoc stripes. A longitudinally striped boundary of Aldoc expression was first identified in the mouse flocculus, and was correlated with the climbing fiber projection pattern and expression of another compartmental marker molecule, heat shock protein 25 (HSP25). As in the rat, the cerebellar nuclei were divided into the rostrodorsal negative and the caudoventral positive portions by distinct projections of Aldoc-positive and negative PC axons in the mouse. Identification of the cerebellar Aldoc stripes in this study, as indicated in sample coronal and horizontal sections as well as in sample surface photos of whole-mount preparations, can be referred to in future experiments.
Collapse
Affiliation(s)
- Hirofumi Fujita
- Department of Systems Neurophysiology, Tokyo Medical and Dental University Graduate School, Bunkyo-ku, Tokyo, Japan
- Systems Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, California, United States of America
- Department of Otolaryngology-Head and Neck Surgery, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Hanako Aoki
- Department of Systems Neurophysiology, Tokyo Medical and Dental University Graduate School, Bunkyo-ku, Tokyo, Japan
| | - Itsuki Ajioka
- Center for Brain Integration Research, Tokyo Medical and Dental University Graduate School, Bunkyo-ku, Tokyo, Japan
| | - Maya Yamazaki
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Manabu Abe
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Arata Oh-Nishi
- Molecular Neuroimaging Program, Molecular Imaging Center, National Institute of Radiological Sciences, Inage-ku, Chiba, Japan
| | - Kenji Sakimura
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Izumi Sugihara
- Department of Systems Neurophysiology, Tokyo Medical and Dental University Graduate School, Bunkyo-ku, Tokyo, Japan
- Center for Brain Integration Research, Tokyo Medical and Dental University Graduate School, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
46
|
c-Jun N-terminal kinase phosphorylation of heterogeneous nuclear ribonucleoprotein K regulates vertebrate axon outgrowth via a posttranscriptional mechanism. J Neurosci 2013; 33:14666-80. [PMID: 24027268 DOI: 10.1523/jneurosci.4821-12.2013] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
c-Jun N-terminal kinase (JNK) mediates cell signaling essential for axon outgrowth, but the associated substrates and underlying mechanisms are poorly understood. We identified in Xenopus laevis embryos a novel posttranscriptional mechanism whereby JNK regulates axonogenesis by phosphorylating a specific site on heterogeneous nuclear ribonucleoprotein K (hnRNP K). Both JNK inhibition and hnRNP K knockdown inhibited axon outgrowth and translation of hnRNP K-regulated cytoskeletal RNAs (tau and neurofilament medium), effects that were alleviated by expressing phosphomimetic, but not phosphodeficient, forms of hnRNP K. Immunohistochemical and biochemical analyses indicated that JNK phosphorylation of hnRNP K occurred within the cytoplasm and was necessary for the translational initiation of hnRNP K-targeted RNAs but not for hnRNP K intracellular localization or RNA binding. Thus, in addition to its known roles in transcription and cytoskeletal organization, JNK acts posttranscriptionally through hnRNP K to regulate translation of proteins crucial for axonogenesis.
Collapse
|
47
|
Liu CH, Chien CL. Molecular cloning and characterization of chicken neuronal intermediate filament protein α-internexin. J Comp Neurol 2013; 521:2147-64. [PMID: 23224860 DOI: 10.1002/cne.23278] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Revised: 09/19/2012] [Accepted: 11/28/2012] [Indexed: 01/20/2023]
Abstract
α-Internexin is one of the neuronal intermediate filament (IF) proteins, which also include low-, middle-, and high-molecular-weight neurofilament (NF) triplet proteins, designated NFL, NFM, and NFH, respectively. The expression of α-internexin occurs in most neurons as they begin differentiation and precedes the expression of the NF triplet proteins in mammals. However, little is known about the gene sequence and physiological function of α-internexin in avians. In this study we describe the molecular cloning of the mRNA sequence encoding the chicken α-internexin (chkINA) protein from embryonic brains. The gene structure and predicted amino acid sequence of chkINA exhibited high similarity to those of its zebrafish, mouse, rat, bovine, and human homologs. Data from transient-transfection experiments show that the filamentous pattern of chkINA was found in transfected cells and colocalized with other endogenous IFs, as demonstrated via immunocytochemistry using a chicken-specific antibody. The expression of chkINA was detected at the early stage of development and increased during the developmental process of the chicken. chkINA was expressed widely in chicken brains and colocalized with NF triplet proteins in neuronal processes, as assessed using immunohistochemistry. We also found that chkINA was expressed abundantly in the developing cerebellum and was the major IF protein in the parallel processes of granule neurons. Thus, we suggest that chkINA is a neuron-specific IF protein that may be a useful marker for studies of chicken brain development.
Collapse
Affiliation(s)
- Chi-Hsiu Liu
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei 10051, Taiwan ROC
| | | |
Collapse
|
48
|
Mitew S, Kirkcaldie MTK, Dickson TC, Vickers JC. Neurites containing the neurofilament-triplet proteins are selectively vulnerable to cytoskeletal pathology in Alzheimer's disease and transgenic mouse models. Front Neuroanat 2013; 7:30. [PMID: 24133416 PMCID: PMC3783838 DOI: 10.3389/fnana.2013.00030] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Accepted: 09/08/2013] [Indexed: 11/17/2022] Open
Abstract
Amyloid-β plaque accumulation in Alzheimer’s disease (AD) is associated with dystrophic neurite (DN) formation and synapse loss in principal neurons, but interneuron pathology is less clearly characterized. We compared the responses of neuronal processes immunoreactive for either neurofilament triplet (NF+) or calretinin (CR+) to fibrillar amyloid (Aβ) plaques in human end-stage and preclinical AD, as well as in APP/PS1 and Tg2576 transgenic mouse AD models. Neurites traversing the Aβ plaque core, edge, or periphery, defined as 50, 100, and 150% of the plaque diameter, respectively, in human AD and transgenic mouse tissue were compared to age-matched human and wild-type mouse controls. The proportion of NF+ neurites exhibiting dystrophic morphology (DN) was significantly larger than the proportion of dystrophic CR+ neurites in both human AD and transgenic mice (p < 0.01). Additionally, the number of NF+, but not CR+, DNs, correlated with Aβ plaque size. We conclude that CR+ interneurons appear to be more resistant than NF+ neurons to AD-mediated cytoskeletal pathology.
Collapse
Affiliation(s)
- Stanislaw Mitew
- Wicking Dementia Research and Education Centre, University of Tasmania Hobart, TAS, Australia ; School of Medicine, University of Tasmania Hobart, TAS, Australia
| | | | | | | |
Collapse
|
49
|
Samantaray S, Knaryan VH, Shields DC, Banik NL. Critical role of calpain in spinal cord degeneration in Parkinson's disease. J Neurochem 2013; 127:880-90. [PMID: 23875735 DOI: 10.1111/jnc.12374] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2013] [Revised: 06/26/2013] [Accepted: 07/05/2013] [Indexed: 11/29/2022]
Abstract
While multiple molecular mechanisms contribute to midbrain nigrostriatal dopaminergic degeneration in Parkinson's disease (PD), the mechanism of damage in non-dopaminergic sites within the central nervous system, including the spinal cord, is not well-understood. Thus, to understand the comprehensive pathophysiology underlying this devastating disease, postmortem spinal cord tissue samples (cervical, thoracic, and lumbar segments) from patients with PD were analyzed compared to age-matched normal subjects or Alzheimer's disease for selective molecular markers of neurodegeneration and inflammation. Distal axonal degeneration, relative abundance of both sensory and motor neuron death, selective loss of ChAT(+) motoneurons, reactive astrogliosis, microgliosis, increased cycloxygenase-2 (Cox-2) expression, and infiltration of T cells were observed in spinal cord of PD patients compared to normal subjects. Biochemical analyses of spinal cord tissues revealed associated inflammatory and proteolytic events (elevated levels of Cox-2, expression and activity of μ- and m-calpain, degradation of axonal neurofilament protein, and concomitantly low levels of endogenous inhibitor - calpastatin) in spinal cord of PD patients. Thus, pathologically upregulated calpain activity in spinal cords of patients with PD may contribute to inflammatory response-mediated neuronal death, leading to motor dysfunction. We proposed calpain over-activation and calpain-calpastatin dysregulation driving in a cascade of inflammatory responses (microglial activation and T cell infiltration) and degenerative pathways culminating in axonal degeneration and neuronal death in spinal cord of Parkinson's disease patients. This may be one of the crucial mechanisms in the degenerative process.
Collapse
Affiliation(s)
- Supriti Samantaray
- Department of Neurosciences, Medical University of South Carolina, 96 Jonathan Lucas Street, Suite 309 CSB, P.O. Box 250606, Charleston, SC, 29425, USA
| | - Varduhi H Knaryan
- Department of Neurosciences, Medical University of South Carolina, 96 Jonathan Lucas Street, Suite 309 CSB, P.O. Box 250606, Charleston, SC, 29425, USA
| | - Donald C Shields
- Department of Neurosurgery, The George Washington University, 2150 Pennsylvania Avenue, NW, Suite 7-420, Washington, DC, 20037, USA
| | - Naren L Banik
- Department of Neurosciences, Medical University of South Carolina, 96 Jonathan Lucas Street, Suite 309 CSB, P.O. Box 250606, Charleston, SC, 29425, USA
| |
Collapse
|
50
|
Abstract
Axon regeneration after damage is widespread in the animal kingdom, and the nematode Caenorhabditis elegans has recently emerged as a tractable model in which to study the genetics and cell biology of axon regrowth in vivo. A key early step in axon regrowth is the conversion of part of a mature axon shaft into a growth cone-like structure, involving coordinated alterations in the microtubule, actin, and neurofilament systems. Recent attention has focused on microtubule dynamics as a determinant of axon-regrowth ability in several organisms. Live imaging studies have begun to reveal how the microtubule cytoskeleton is remodeled after axon injury, as well as the regulatory pathways involved. The dual leucine zipper kinase family of mixed-lineage kinases has emerged as a critical sensor of axon damage and plays a key role in regulating microtubule dynamics in the damaged axon.
Collapse
Affiliation(s)
- Andrew D Chisholm
- Division of Biological Sciences, Section of Neurobiology, University of California, San Diego, La Jolla, California 92093;
| |
Collapse
|