1
|
Dapper K, Wolpert SM, Schirmer J, Fink S, Gaudrain E, Başkent D, Singer W, Verhulst S, Braun C, Dalhoff E, Rüttiger L, Munk MHJ, Knipper M. Age dependent deficits in speech recognition in quiet and noise are reflected in MGB activity and cochlear onset coding. Neuroimage 2025; 305:120958. [PMID: 39622462 DOI: 10.1016/j.neuroimage.2024.120958] [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: 05/29/2024] [Revised: 11/25/2024] [Accepted: 11/26/2024] [Indexed: 12/21/2024] Open
Abstract
The slowing and reduction of auditory responses in the brain are recognized side effects of increased pure tone thresholds, impaired speech recognition, and aging. However, it remains controversial whether central slowing is primarily linked to brain processes as atrophy, or is also associated with the slowing of temporal neural processing from the periphery. Here we analyzed electroencephalogram (EEG) responses that most likely reflect medial geniculate body (MGB) responses to passive listening of phonemes in 80 subjects ranging in age from 18 to 76 years, in whom the peripheral auditory responses had been analyzed in detail (Schirmer et al., 2024). We observed that passive listening to vowels and phonemes, specifically designed to rely on either temporal fine structure (TFS) for frequencies below the phase locking limit (<1500 Hz), or on the temporal envelope (TENV) for frequencies above phase locking limit, entrained lower or higher neural EEG responses. While previous views predict speech content, particular in noise to be encoded through TENV, here a decreasing phoneme-induced EEG amplitude over age in response to phonemes relying on TENV coding could also be linked to poorer speech-recognition thresholds in quiet. In addition, increased phoneme-evoked EEG delay could be correlated with elevated extended high-frequency threshold (EHF) for phoneme changes that relied on TFS and TENV coding. This may suggest a role of pure-tone threshold averages (PTA) of EHF for TENV and TFS beyond sound localization that is reflected in likely MGB delays. When speech recognition thresholds were normalized for pure-tone thresholds, however, the EEG amplitudes remained insignificant, and thereby became independent of age. Under these conditions, poor speech recognition in quiet was found together with a delay in EEG response for phonemes that relied on TFS coding, while poor speech recognition in ipsilateral noise was observed as a trend of shortened EEG delays for phonemes that relied on TENV coding. Based on previous analyses performed in these same subjects, elevated thresholds in extended high-frequency regions were linked to cochlear synaptopathy and auditory brainstem delays. Also, independent of hearing loss, poor speech-performing groups in quiet or with ipsilateral noise during TFS or TENV coding could be linked to lower or better outer hair cell performance and delayed or steeper auditory nerve responses at stimulus onset. The amplitude and latency of MGB responses to phonemes requiring TFS or TENV coding, dependent or independent of hearing loss, may thus be a new predictor of poor speech recognition in quiet and ipsilateral noise that links deficits in synchronicity at stimulus onset to neocortical activity. Amplitudes and delays of speech EEG responses to syllables should be reconsidered for future hearing-aid studies.
Collapse
Affiliation(s)
- Konrad Dapper
- Department of Otolaryngology, Head and Neck, University of Tübingen, Tübingen 72076, Germany; Department of Biology, Technical University 64287 Darmstadt, Darmstadt, Germany
| | - Stephan M Wolpert
- Department of Otolaryngology, Head and Neck, University of Tübingen, Tübingen 72076, Germany
| | - Jakob Schirmer
- Department of Otolaryngology, Head and Neck, University of Tübingen, Tübingen 72076, Germany
| | - Stefan Fink
- Department of Otolaryngology, Head and Neck, University of Tübingen, Tübingen 72076, Germany
| | - Etienne Gaudrain
- Lyon Neuroscience Research Center, Université Claude Bernard Lyon 1, CNRS UMR5292, INSERM U1028, Center Hospitalier Le Vinatier -Bâtiment 462-Neurocampus, 95 boulevard Pinel, Lyon, France
| | - Deniz Başkent
- Department of Otorhinolaryngology, University Medical Center Groningen (UMCG), Hanzeplein 1, BB21, Groningen 9700RB, the Netherlands
| | - Wibke Singer
- Department of Otolaryngology, Head and Neck, University of Tübingen, Tübingen 72076, Germany
| | - Sarah Verhulst
- Department of Information Technology, Ghent University, Zwijnaarde 9052, Belgium
| | - Christoph Braun
- MEG-Center, University of Tübingen, Tübingen 72076, Germany; HIH, Hertie Institute for Clinical Brain Research, Tübingen 72076, Germany; CIMeC, Center for Mind and Brain Research, University of Trento, Rovereto 38068, Italy
| | - Ernst Dalhoff
- Department of Otolaryngology, Head and Neck, University of Tübingen, Tübingen 72076, Germany
| | - Lukas Rüttiger
- Department of Otolaryngology, Head and Neck, University of Tübingen, Tübingen 72076, Germany
| | - Matthias H J Munk
- Department of Otolaryngology, Head and Neck, University of Tübingen, Tübingen 72076, Germany; Department of Biology, Technical University 64287 Darmstadt, Darmstadt, Germany
| | - Marlies Knipper
- Department of Otolaryngology, Head and Neck, University of Tübingen, Tübingen 72076, Germany.
| |
Collapse
|
2
|
Lian X, Bai Y, Du P, Jing Z, Gao J, Liu F, Hu J, Xi Y. Causal influences of testosterone on brain structure change rate: A sex-stratified Mendelian randomization study. J Steroid Biochem Mol Biol 2025; 245:106629. [PMID: 39481491 DOI: 10.1016/j.jsbmb.2024.106629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 10/26/2024] [Accepted: 10/29/2024] [Indexed: 11/02/2024]
Abstract
The impact of testosterone levels on changes in brain structure has been reported. However, it is still unclear which specific brain region could be affected. This study approached Mendelian randomization method to reveal the causal relationship between testosterone levels and the rate of longitudinal structural changes in the brain. The testosterone-related GWAS data were determined from 425,097 European participants. The GWAS data on the rate of longitudinal structural changes in the brain came from the ENIGMA consortium, which included 15,640 all-age participants from 40 longitudinal cohorts. The inverse variance weighted was considered as the main estimate, MR Egger and weighted median methods were used to supplement IVW. A positive correlation was found between total testosterone levels and bioavailable testosterone levels in women and age-independent longitudinal changes in cerebral WM and surface area. The sex hormone-binding globulin levels were found a negative correlation with age-dependent longitudinal structural changes of cortical GM in men. Additionally, we also found that the bioavailable testosterone level in males was negatively associated with the quadratic age-dependent longitudinal change rate in the globus pallidum. We also found estradiol levels and sex hormone-binding globulin levels were negatively associated with the quadratic age-dependent longitudinal change rate of total brain in men. Moreover, we found a positive correlation between total testosterone levels and linear age-dependent longitudinal changes in the hippocampus in both males and females. The testosterone levels in different genders may have varying degrees of causal effects on the structural changes of brain regions. These findings provide evidence for the influence of the brain glandular axis on brain structure, particularly during female brain development.
Collapse
Affiliation(s)
- Xin Lian
- Department of Obstetrics and Gynecology, People's Hospital of Linyi County, Yuncheng Central Hospital of Shanxi Medical University, 1125 Fuxi Street, Yuncheng 044100, China
| | - Yaqi Bai
- School of Clinical Medicine, Shanxi Medical University, 56 Xinjian South Road, Taiyuan 030000, China
| | - Pengyang Du
- Department of Neurology, Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan 030001, China
| | - Zhinan Jing
- School of Clinical Medicine, Shanxi Medical University, 56 Xinjian South Road, Taiyuan 030000, China
| | - Jimi Gao
- Department of Obstetrics and Gynecology, Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan 030001, China
| | - Fan Liu
- Department of Neurology, Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan 030001, China
| | - Jingjing Hu
- Department of Obstetrics and Gynecology, Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan 030001, China.
| | - Yujia Xi
- Department of Urology, Second Hospital of Shanxi Medical University, Male Reproductive Medicine Center, 382 Wuyi Road, Taiyuan 030001, China.
| |
Collapse
|
3
|
Ding EA, Kumar S. Neurofilament Biophysics: From Structure to Biomechanics. Mol Biol Cell 2024; 35:re1. [PMID: 38598299 PMCID: PMC11151108 DOI: 10.1091/mbc.e23-11-0438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 03/25/2024] [Accepted: 04/04/2024] [Indexed: 04/12/2024] Open
Abstract
Neurofilaments (NFs) are multisubunit, neuron-specific intermediate filaments consisting of a 10-nm diameter filament "core" surrounded by a layer of long intrinsically disordered protein (IDP) "tails." NFs are thought to regulate axonal caliber during development and then stabilize the mature axon, with NF subunit misregulation, mutation, and aggregation featuring prominently in multiple neurological diseases. The field's understanding of NF structure, mechanics, and function has been deeply informed by a rich variety of biochemical, cell biological, and mouse genetic studies spanning more than four decades. These studies have contributed much to our collective understanding of NF function in axonal physiology and disease. In recent years, however, there has been a resurgence of interest in NF subunit proteins in two new contexts: as potential blood- and cerebrospinal fluid-based biomarkers of neuronal damage, and as model IDPs with intriguing properties. Here, we review established principles and more recent discoveries in NF structure and function. Where possible, we place these findings in the context of biophysics of NF assembly, interaction, and contributions to axonal mechanics.
Collapse
Affiliation(s)
- Erika A. Ding
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA 94720
| | - Sanjay Kumar
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA 94720
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA 94158
| |
Collapse
|
4
|
Shahim P, Norato G, Sinaii N, Zetterberg H, Blennow K, Chan L, Grunseich C. Neurofilaments in Sporadic and Familial Amyotrophic Lateral Sclerosis: A Systematic Review and Meta-Analysis. Genes (Basel) 2024; 15:496. [PMID: 38674431 PMCID: PMC11050235 DOI: 10.3390/genes15040496] [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: 03/03/2024] [Revised: 03/30/2024] [Accepted: 04/02/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND Neurofilament proteins have been implicated to be altered in amyotrophic lateral sclerosis (ALS). The objectives of this study were to assess the diagnostic and prognostic utility of neurofilaments in ALS. METHODS Studies were conducted in electronic databases (PubMed/MEDLINE, Embase, Web of Science, and Cochrane CENTRAL) from inception to 17 August 2023, and investigated neurofilament light (NfL) or phosphorylated neurofilament heavy chain (pNfH) in ALS. The study design, enrolment criteria, neurofilament concentrations, test accuracy, relationship between neurofilaments in cerebrospinal fluid (CSF) and blood, and clinical outcome were recorded. The protocol was registered with PROSPERO, CRD42022376939. RESULTS Sixty studies with 8801 participants were included. Both NfL and pNfH measured in CSF showed high sensitivity and specificity in distinguishing ALS from disease mimics. Both NfL and pNfH measured in CSF correlated with their corresponding levels in blood (plasma or serum); however, there were stronger correlations between CSF NfL and blood NfL. NfL measured in blood exhibited high sensitivity and specificity in distinguishing ALS from controls. Both higher levels of NfL and pNfH either measured in blood or CSF were correlated with more severe symptoms as assessed by the ALS Functional Rating Scale Revised score and with a faster disease progression rate; however, only blood NfL levels were associated with shorter survival. DISCUSSION Both NfL and pNfH measured in CSF or blood show high diagnostic utility and association with ALS functional scores and disease progression, while CSF NfL correlates strongly with blood (either plasma or serum) and is also associated with survival, supporting its use in clinical diagnostics and prognosis. Future work must be conducted in a prospective manner with standardized bio-specimen collection methods and analytical platforms, further improvement in immunoassays for quantification of pNfH in blood, and the identification of cut-offs across the ALS spectrum and controls.
Collapse
Affiliation(s)
- Pashtun Shahim
- Rehabilitation Medicine Department, National Institutes of Health (NIH) Clinical Center, Bethesda, MD 20892, USA;
- National Institutes of Neurological Disorders and Stroke, NIH, Bethesda, MD 20892, USA; (G.N.); (C.G.)
- Department of Neurology, MedStar Georgetown University Hospital, Washington, DC 20007, USA
- The Military Traumatic Brain Injury Initiative (MTBI2), Bethesda, MD 20814, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817, USA
| | - Gina Norato
- National Institutes of Neurological Disorders and Stroke, NIH, Bethesda, MD 20892, USA; (G.N.); (C.G.)
| | - Ninet Sinaii
- Biostatistics and Clinical Epidemiology Service, NIH, Bethesda, MD 20892, USA;
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 431 41 Molndal, Sweden; (H.Z.); (K.B.)
- Clinical Neurochemistry Laboratory, Sahglrenska University Hospital, 431 41 Molndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
- UK Dementia Research Institute at UCL, London WC1E 6BT, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong 518172, China
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53792, USA
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 431 41 Molndal, Sweden; (H.Z.); (K.B.)
- Clinical Neurochemistry Laboratory, Sahglrenska University Hospital, 431 41 Molndal, Sweden
| | - Leighton Chan
- Rehabilitation Medicine Department, National Institutes of Health (NIH) Clinical Center, Bethesda, MD 20892, USA;
| | - Christopher Grunseich
- National Institutes of Neurological Disorders and Stroke, NIH, Bethesda, MD 20892, USA; (G.N.); (C.G.)
| |
Collapse
|
5
|
Zhou Y, He W, Hou W, Zhu Y. Pianno: a probabilistic framework automating semantic annotation for spatial transcriptomics. Nat Commun 2024; 15:2848. [PMID: 38565531 PMCID: PMC11271244 DOI: 10.1038/s41467-024-47152-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 03/20/2024] [Indexed: 04/04/2024] Open
Abstract
Spatial transcriptomics has revolutionized the study of gene expression within tissues, while preserving spatial context. However, annotating spatial spots' biological identity remains a challenge. To tackle this, we introduce Pianno, a Bayesian framework automating structural semantics annotation based on marker genes. Comprehensive evaluations underscore Pianno's remarkable prowess in precisely annotating a wide array of spatial semantics, ranging from diverse anatomical structures to intricate tumor microenvironments, as well as in estimating cell type distributions, across data generated from various spatial transcriptomics platforms. Furthermore, Pianno, in conjunction with clustering approaches, uncovers a region- and species-specific excitatory neuron subtype in the deep layer 3 of the human neocortex, shedding light on cellular evolution in the human neocortex. Overall, Pianno equips researchers with a robust and efficient tool for annotating diverse biological structures, offering new perspectives on spatial transcriptomics data.
Collapse
Affiliation(s)
- Yuqiu Zhou
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science and Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Wei He
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science and Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Weizhen Hou
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science and Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Ying Zhu
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science and Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
6
|
López-Erauskin J, Bravo-Hernandez M, Presa M, Baughn MW, Melamed Z, Beccari MS, Agra de Almeida Quadros AR, Arnold-Garcia O, Zuberi A, Ling K, Platoshyn O, Niño-Jara E, Ndayambaje IS, McAlonis-Downes M, Cabrera L, Artates JW, Ryan J, Hermann A, Ravits J, Bennett CF, Jafar-Nejad P, Rigo F, Marsala M, Lutz CM, Cleveland DW, Lagier-Tourenne C. Stathmin-2 loss leads to neurofilament-dependent axonal collapse driving motor and sensory denervation. Nat Neurosci 2024; 27:34-47. [PMID: 37996528 PMCID: PMC10842032 DOI: 10.1038/s41593-023-01496-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Accepted: 10/16/2023] [Indexed: 11/25/2023]
Abstract
The mRNA transcript of the human STMN2 gene, encoding for stathmin-2 protein (also called SCG10), is profoundly impacted by TAR DNA-binding protein 43 (TDP-43) loss of function. The latter is a hallmark of several neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS). Using a combination of approaches, including transient antisense oligonucleotide-mediated suppression, sustained shRNA-induced depletion in aging mice, and germline deletion, we show that stathmin-2 has an important role in the establishment and maintenance of neurofilament-dependent axoplasmic organization that is critical for preserving the caliber and conduction velocity of myelinated large-diameter axons. Persistent stathmin-2 loss in adult mice results in pathologies found in ALS, including reduced interneurofilament spacing, axonal caliber collapse that drives tearing within outer myelin layers, diminished conduction velocity, progressive motor and sensory deficits, and muscle denervation. These findings reinforce restoration of stathmin-2 as an attractive therapeutic approach for ALS and other TDP-43-dependent neurodegenerative diseases.
Collapse
Affiliation(s)
- Jone López-Erauskin
- Ludwig Institute and Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA, USA
| | - Mariana Bravo-Hernandez
- Department of Anesthesiology and Stem Cell Program and Institute for Genomic Medicine, University of California at San Diego, La Jolla, CA, USA
- Ionis Pharmaceuticals Inc., Carlsbad, CA, USA
| | | | - Michael W Baughn
- Ludwig Institute and Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA, USA
| | - Ze'ev Melamed
- Ludwig Institute and Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA, USA
- Department of Medical Neurobiology, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Melinda S Beccari
- Ludwig Institute and Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA, USA
| | - Ana Rita Agra de Almeida Quadros
- Department of Neurology, The Sean M. Healey and AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Olatz Arnold-Garcia
- Ludwig Institute and Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA, USA
- Department of Neurosciences, Biodonostia Health Research Institute, San Sebastián, Spain
- CIBERNED, ISCIII (CIBER, Carlos III Institute, Spanish Ministry of Sciences and Innovation), Madrid, Spain
| | | | - Karen Ling
- Ionis Pharmaceuticals Inc., Carlsbad, CA, USA
| | - Oleksandr Platoshyn
- Department of Anesthesiology and Stem Cell Program and Institute for Genomic Medicine, University of California at San Diego, La Jolla, CA, USA
| | - Elkin Niño-Jara
- Department of Anesthesiology and Stem Cell Program and Institute for Genomic Medicine, University of California at San Diego, La Jolla, CA, USA
| | - I Sandra Ndayambaje
- Department of Neurology, The Sean M. Healey and AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Melissa McAlonis-Downes
- Ludwig Institute and Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA, USA
| | - Larissa Cabrera
- Ludwig Institute and Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA, USA
| | - Jonathan W Artates
- Ludwig Institute and Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA, USA
| | | | - Anita Hermann
- Department of Neurosciences, School of Medicine, University of California at San Diego, La Jolla, CA, USA
| | - John Ravits
- Department of Neurosciences, School of Medicine, University of California at San Diego, La Jolla, CA, USA
| | | | | | - Frank Rigo
- Ionis Pharmaceuticals Inc., Carlsbad, CA, USA
| | - Martin Marsala
- Department of Anesthesiology and Stem Cell Program and Institute for Genomic Medicine, University of California at San Diego, La Jolla, CA, USA
| | | | - Don W Cleveland
- Ludwig Institute and Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA, USA.
| | - Clotilde Lagier-Tourenne
- Department of Neurology, The Sean M. Healey and AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Broad Institute of Harvard and MIT, Cambridge, MA, USA.
| |
Collapse
|
7
|
Yadav A, Matson KJE, Li L, Hua I, Petrescu J, Kang K, Alkaslasi MR, Lee DI, Hasan S, Galuta A, Dedek A, Ameri S, Parnell J, Alshardan MM, Qumqumji FA, Alhamad SM, Wang AP, Poulen G, Lonjon N, Vachiery-Lahaye F, Gaur P, Nalls MA, Qi YA, Maric D, Ward ME, Hildebrand ME, Mery PF, Bourinet E, Bauchet L, Tsai EC, Phatnani H, Le Pichon CE, Menon V, Levine AJ. A cellular taxonomy of the adult human spinal cord. Neuron 2023; 111:328-344.e7. [PMID: 36731429 PMCID: PMC10044516 DOI: 10.1016/j.neuron.2023.01.007] [Citation(s) in RCA: 87] [Impact Index Per Article: 43.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 11/30/2022] [Accepted: 01/11/2023] [Indexed: 02/04/2023]
Abstract
The mammalian spinal cord functions as a community of cell types for sensory processing, autonomic control, and movement. While animal models have advanced our understanding of spinal cellular diversity, characterizing human biology directly is important to uncover specialized features of basic function and human pathology. Here, we present a cellular taxonomy of the adult human spinal cord using single-nucleus RNA sequencing with spatial transcriptomics and antibody validation. We identified 29 glial clusters and 35 neuronal clusters, organized principally by anatomical location. To demonstrate the relevance of this resource to human disease, we analyzed spinal motoneurons, which degenerate in amyotrophic lateral sclerosis (ALS) and other diseases. We found that compared with other spinal neurons, human motoneurons are defined by genes related to cell size, cytoskeletal structure, and ALS, suggesting a specialized molecular repertoire underlying their selective vulnerability. We include a web resource to facilitate further investigations into human spinal cord biology.
Collapse
Affiliation(s)
- Archana Yadav
- Department of Neurology, Center for Translational and Computational Neuroimmunology, Columbia University, New York, NY, USA
| | - Kaya J E Matson
- Spinal Circuits and Plasticity Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA; Johns Hopkins University Department of Biology, Baltimore, MD 21218, USA
| | - Li Li
- Spinal Circuits and Plasticity Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Isabelle Hua
- Spinal Circuits and Plasticity Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Joana Petrescu
- Department of Neurology, Center for Translational and Computational Neuroimmunology, Columbia University, New York, NY, USA; Center for Genomics of Neurodegenerative Disease, New York Genome Center, New York, NY, USA
| | - Kristy Kang
- Department of Neurology, Center for Translational and Computational Neuroimmunology, Columbia University, New York, NY, USA; Center for Genomics of Neurodegenerative Disease, New York Genome Center, New York, NY, USA
| | - Mor R Alkaslasi
- Unit on the Development of Neurodegeneration, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, USA; Department of Neuroscience, Brown University, Providence, RI, USA
| | - Dylan I Lee
- Department of Neurology, Center for Translational and Computational Neuroimmunology, Columbia University, New York, NY, USA
| | - Saadia Hasan
- Inherited Neurodegenerative Diseases Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Ahmad Galuta
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Annemarie Dedek
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada; Department of Neuroscience, Carleton University, Ottawa, ON, Canada
| | - Sara Ameri
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Jessica Parnell
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada; Department of Neuroscience, Carleton University, Ottawa, ON, Canada
| | | | | | - Saud M Alhamad
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Alick Pingbei Wang
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Gaetan Poulen
- Department of Neurosurgery, Gui de Chauliac Hospital, and Donation and Transplantation Coordination Unit, Montpellier University Medical Center, Montpellier, France
| | - Nicolas Lonjon
- Department of Neurosurgery, Gui de Chauliac Hospital, and Donation and Transplantation Coordination Unit, Montpellier University Medical Center, Montpellier, France
| | - Florence Vachiery-Lahaye
- Department of Neurosurgery, Gui de Chauliac Hospital, and Donation and Transplantation Coordination Unit, Montpellier University Medical Center, Montpellier, France
| | - Pallavi Gaur
- Department of Neurology, Center for Translational and Computational Neuroimmunology, Columbia University, New York, NY, USA
| | - Mike A Nalls
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA; Center for Alzheimer's and Related Dementias, National Institutes of Health, Bethesda, MD, USA; Data Tecnica International LLC, Glen Echo, MD, USA
| | - Yue A Qi
- Center for Alzheimer's and Related Dementias, National Institutes of Health, Bethesda, MD, USA
| | - Dragan Maric
- Flow and Imaging Cytometry Core Facility, National Institute of Neurological Disorders and Stroke; Bethesda, MD, USA
| | - Michael E Ward
- Inherited Neurodegenerative Diseases Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Michael E Hildebrand
- Inherited Neurodegenerative Diseases Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA; Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Pierre-Francois Mery
- Institute of Functional Genomics, Montpellier University, CNRS, INSERM, Montpellier, France
| | - Emmanuel Bourinet
- Institute of Functional Genomics, Montpellier University, CNRS, INSERM, Montpellier, France
| | - Luc Bauchet
- Department of Neurosurgery, Gui de Chauliac Hospital, and Donation and Transplantation Coordination Unit, Montpellier University Medical Center, Montpellier, France; Institute of Functional Genomics, Montpellier University, CNRS, INSERM, Montpellier, France
| | - Eve C Tsai
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Hemali Phatnani
- Department of Neurology, Center for Translational and Computational Neuroimmunology, Columbia University, New York, NY, USA; Center for Genomics of Neurodegenerative Disease, New York Genome Center, New York, NY, USA
| | - Claire E Le Pichon
- Unit on the Development of Neurodegeneration, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, USA
| | - Vilas Menon
- Department of Neurology, Center for Translational and Computational Neuroimmunology, Columbia University, New York, NY, USA.
| | - Ariel J Levine
- Spinal Circuits and Plasticity Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA.
| |
Collapse
|
8
|
Petzold A. The 2022 Lady Estelle Wolfson lectureship on neurofilaments. J Neurochem 2022; 163:179-219. [PMID: 35950263 PMCID: PMC9826399 DOI: 10.1111/jnc.15682] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/19/2022] [Accepted: 07/21/2022] [Indexed: 01/11/2023]
Abstract
Neurofilament proteins (Nf) have been validated and established as a reliable body fluid biomarker for neurodegenerative pathology. This review covers seven Nf isoforms, Nf light (NfL), two splicing variants of Nf medium (NfM), two splicing variants of Nf heavy (NfH),α -internexin (INA) and peripherin (PRPH). The genetic and epigenetic aspects of Nf are discussed as relevant for neurodegenerative diseases and oncology. The comprehensive list of mutations for all Nf isoforms covers Amyotrophic Lateral Sclerosis, Charcot-Marie Tooth disease, Spinal muscular atrophy, Parkinson Disease and Lewy Body Dementia. Next, emphasis is given to the expanding field of post-translational modifications (PTM) of the Nf amino acid residues. Protein structural aspects are reviewed alongside PTMs causing neurodegenerative pathology and human autoimmunity. Molecular visualisations of NF PTMs, assembly and stoichiometry make use of Alphafold2 modelling. The implications for Nf function on the cellular level and axonal transport are discussed. Neurofilament aggregate formation and proteolytic breakdown are reviewed as relevant for biomarker tests and disease. Likewise, Nf stoichiometry is reviewed with regard to in vitro experiments and as a compensatory mechanism in neurodegeneration. The review of Nf across a spectrum of 87 diseases from all parts of medicine is followed by a critical appraisal of 33 meta-analyses on Nf body fluid levels. The review concludes with considerations for clinical trial design and an outlook for future research.
Collapse
Affiliation(s)
- Axel Petzold
- Department of NeurodegenerationQueen Square Insitute of Neurology, UCLLondonUK
| |
Collapse
|
9
|
Molotsky E, Liu Y, Lieberman AP, Merry DE. Neuromuscular junction pathology is correlated with differential motor unit vulnerability in spinal and bulbar muscular atrophy. Acta Neuropathol Commun 2022; 10:97. [PMID: 35791011 PMCID: PMC9258097 DOI: 10.1186/s40478-022-01402-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 06/23/2022] [Indexed: 11/10/2022] Open
Abstract
Spinal and bulbar muscular atrophy (SBMA) is an X-linked, neuromuscular neurodegenerative disease for which there is no cure. The disease is characterized by a selective decrease in fast-muscle power (e.g., tongue pressure, grip strength) accompanied by a selective loss of fast-twitch muscle fibers. However, the relationship between neuromuscular junction (NMJ) pathology and fast-twitch motor unit vulnerability has yet to be explored. In this study, we used a cross-model comparison of two mouse models of SBMA to evaluate neuromuscular junction pathology, glycolytic-to-oxidative fiber-type switching, and cytoskeletal alterations in pre- and postsynaptic termini of tibialis anterior (TA), gastrocnemius, and soleus hindlimb muscles. We observed significantly increased NMJ and myofiber pathology in fast-twitch, glycolytic motor units of the TA and gastrocnemius compared to slow-twitch, oxidative motor units of the soleus, as seen by decreased pre- and post-synaptic membrane area, decreased pre- and post-synaptic membrane colocalization, increased acetylcholine receptor compactness, a decrease in endplate area and complexity, and deficits in neurofilament heavy chain. Our data also show evidence for metabolic dysregulation and myofiber atrophy that correlate with severity of NMJ pathology. We propose a model in which the dynamic communicative relationship between the motor neuron and muscle, along with the developmental subtype of the muscle, promotes motor unit subtype specific vulnerability, metabolic alterations, and NMJ pathology.
Collapse
Affiliation(s)
- Elana Molotsky
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Jefferson Alumni Hall, Rm. 411E, Philadelphia, PA, 19107, USA
| | - Yuhong Liu
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Jefferson Alumni Hall, Rm. 411E, Philadelphia, PA, 19107, USA
| | - Andrew P Lieberman
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Diane E Merry
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Jefferson Alumni Hall, Rm. 411E, Philadelphia, PA, 19107, USA.
| |
Collapse
|
10
|
The Neuromuscular Junction: Roles in Aging and Neuromuscular Disease. Int J Mol Sci 2021; 22:ijms22158058. [PMID: 34360831 PMCID: PMC8347593 DOI: 10.3390/ijms22158058] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 07/20/2021] [Accepted: 07/22/2021] [Indexed: 02/07/2023] Open
Abstract
The neuromuscular junction (NMJ) is a specialized synapse that bridges the motor neuron and the skeletal muscle fiber and is crucial for conversion of electrical impulses originating in the motor neuron to action potentials in the muscle fiber. The consideration of contributing factors to skeletal muscle injury, muscular dystrophy and sarcopenia cannot be restricted only to processes intrinsic to the muscle, as data show that these conditions incur denervation-like findings, such as fragmented NMJ morphology and corresponding functional changes in neuromuscular transmission. Primary defects in the NMJ also influence functional loss in motor neuron disease, congenital myasthenic syndromes and myasthenia gravis, resulting in skeletal muscle weakness and heightened fatigue. Such findings underscore the role that the NMJ plays in neuromuscular performance. Regardless of cause or effect, functional denervation is now an accepted consequence of sarcopenia and muscle disease. In this short review, we provide an overview of the pathologic etiology, symptoms, and therapeutic strategies related to the NMJ. In particular, we examine the role of the NMJ as a disease modifier and a potential therapeutic target in neuromuscular injury and disease.
Collapse
|
11
|
Jia Z, Li Y. A possible mechanism for neurofilament slowing down in myelinated axon: Phosphorylation-induced variation of NF kinetics. PLoS One 2021; 16:e0247656. [PMID: 33711034 PMCID: PMC7954336 DOI: 10.1371/journal.pone.0247656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 02/10/2021] [Indexed: 11/18/2022] Open
Abstract
Neurofilaments(NFs) are the most abundant intermediate filaments that make up the inner volume of axon, with possible phosphorylation on their side arms, and their slow axonal transport by molecular motors along microtubule tracks in a “stop-and-go” manner with rapid, intermittent and bidirectional motion. The kinetics of NFs and morphology of axon are dramatically different between myelinate internode and unmyelinated node of Ranvier. The NFs in the node transport as 7.6 times faster as in the internode, and the distribution of NFs population in the internode is 7.6 folds as much as in the node of Ranvier. We hypothesize that the phosphorylation of NFs could reduce the on-track rate and slow down their transport velocity in the internode. By modifying the ‘6-state’ model with (a) an extra phosphorylation kinetics to each six state and (b) construction a new ‘8-state’ model in which NFs at off-track can be phosphorylated and have smaller on-track rate, our model and simulation demonstrate that the phosphorylation-induced decrease of on-track rate could slow down the NFs average velocity and increase the axonal caliber. The degree of phosphorylation may indicate the extent of velocity reduction. The Continuity equation used in our paper predicts that the ratio of NFs population is inverse proportional to the ratios of average velocity of NFs between node of Ranvier and internode. We speculate that the myelination of axon could increase the level of phosphorylation of NF side arms, and decrease the possibility of NFs to get on-track of microtubules, therefore slow down their transport velocity. In summary, our work provides a potential mechanism for understanding the phosphorylation kinetics of NFs in regulating their transport and morphology of axon in myelinated axons, and the different kinetics of NFs between node and internode.
Collapse
Affiliation(s)
- Zelin Jia
- School of Systems Science, Beijing Normal University, Beijing, China
| | - Yinyun Li
- School of Systems Science, Beijing Normal University, Beijing, China
- * E-mail:
| |
Collapse
|
12
|
Charvet CJ, Palani A, Kabaria P, Takahashi E. Evolution of Brain Connections: Integrating Diffusion MR Tractography With Gene Expression Highlights Increased Corticocortical Projections in Primates. Cereb Cortex 2020; 29:5150-5165. [PMID: 30927350 DOI: 10.1093/cercor/bhz054] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 02/20/2019] [Indexed: 12/19/2022] Open
Abstract
Diffusion MR tractography permits investigating the 3D structure of cortical pathways as interwoven paths across the entire brain. We use high-resolution scans from diffusion spectrum imaging and high angular resolution diffusion imaging to investigate the evolution of cortical pathways within the euarchontoglire (i.e., primates, rodents) lineage. More specifically, we compare cortical fiber pathways between macaques (Macaca mulatta), marmosets (Callithrix jachus), and rodents (mice, Mus musculus). We integrate these observations with comparative analyses of Neurofilament heavy polypeptide (NEFH) expression across the cortex of mice and primates. We chose these species because their phylogenetic position serves to trace the early evolutionary history of the human brain. Our comparative analysis from diffusion MR tractography, cortical white matter scaling, and NEFH expression demonstrates that the examined primates deviate from mice in possessing increased long-range cross-cortical projections, many of which course across the anterior to posterior axis of the cortex. Our study shows that integrating gene expression data with diffusion MR data is an effective approach in identifying variation in connectivity patterns between species. The expansion of corticocortical pathways and increased anterior to posterior cortical integration can be traced back to an extension of neurogenetic schedules during development in primates.
Collapse
Affiliation(s)
| | - Arthi Palani
- Division of Newborn Medicine, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA 02215, USA.,Medical Sciences in the College of Arts and Sciences, Boston University, Boston, MA 02215, USA
| | - Priya Kabaria
- Division of Newborn Medicine, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA 02215, USA.,Department of Behavioral Neuroscience, Northeastern University, Boston, MA 02115, USA
| | - Emi Takahashi
- Division of Newborn Medicine, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
13
|
Huang P, Chen X, Hu X, Zhou Q, Lin L, Jiang S, Fu H, Xiong Y, Zeng H, Fang M, Chen C, Deng Y. Experimentally Induced Sepsis Causes Extensive Hypomyelination in the Prefrontal Cortex and Hippocampus in Neonatal Rats. Neuromolecular Med 2020; 22:420-436. [PMID: 32638208 DOI: 10.1007/s12017-020-08602-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Accepted: 06/17/2020] [Indexed: 02/05/2023]
Abstract
Neonatal sepsis is associated with cognitive deficit in the later life. Axonal myelination plays a pivotal role in neurotransmission and formation of learning and memory. This study aimed to explore if systemic lipopolysaccharide (LPS) injection would induce hypomyelination in the prefrontal cortex and hippocampus in developing septic neonatal rats. Sprague-Dawley rats (1-day old) were injected with LPS (1 mg/kg) intraperitoneally. By electron microscopy, axonal hypomyelination was evident in the subcortical white matter and hippocampus. The expression of myelin proteins including CNPase, MBP, PLP and MAG was downregulated in both areas of the brain at 7, 14 and 28 days after LPS injection. The frequency of MBP and PLP-positive oligodendrocyte was significantly reduced using in situ hybridization in the cerebral cortex and hippocampus at the corresponding time points after LPS injection, whereas the expression of NG2 and PDGFRα was noticeably increased. In tandem with this was reduction of Olig1 and Olig2 expressions which are involved in differentiation/maturation of OPCs. Expression of NFL, NFM, and NFH was significantly downregulated, indicating that axon development was disrupted after LPS injection. Morris Water Maze behavioral test, Open field test, Rotarod test, and Pole test were used to evaluate neurological behaviors of 28 days rats. The rats in the LPS group showed the impairment of motor coordination, balance, memory, and learning ability and represented bradykinesia and anxiety-like behavior. The present results suggest that following systemic LPS injection, differentiation/maturation of OPCs was affected which may be attributed to the inhibition of transcription factors Olig1 and Olig2 expression resulting in impairment to axonal development. It is suggested that this would ultimately lead to axonal hypomyelination in the prefrontal cortex and hippocampus, which may be associated with neurological deficits in later life.
Collapse
Affiliation(s)
- Peixian Huang
- Department of Critical Care and Emergency, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, China
| | - Xuan Chen
- Department of Critical Care and Emergency, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, China
- Shantou University Medical College, Shantou, 515063, Guangdong, China
| | - Xiaoli Hu
- Department of Operative Dentistry and Endodontics, Guanghua School and Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, 510055, Guangdong, China
| | - Qiuping Zhou
- Department of Critical Care and Emergency, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, China
- South China University of Technology, Guangzhou, 510641, Guangdong, China
| | - Lanfen Lin
- Guangdong Second Provincial General Hospital, Guangzhou, 510317, Guangdong, China
| | - Shuqi Jiang
- Department of Critical Care and Emergency, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, China
- South China University of Technology, Guangzhou, 510641, Guangdong, China
| | - Hui Fu
- Wuhan University School of Basic Medical Sciences, Wuhan, 430072, Hubei, China
| | - Yajie Xiong
- Wuhan University School of Basic Medical Sciences, Wuhan, 430072, Hubei, China
| | - Hongke Zeng
- Department of Critical Care and Emergency, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, China
| | - Ming Fang
- Department of Critical Care and Emergency, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, China
| | - Chunbo Chen
- Department of Intensive Care Unit of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China.
| | - Yiyu Deng
- Department of Critical Care and Emergency, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, China.
| |
Collapse
|
14
|
Goethals S, Brette R. Theoretical relation between axon initial segment geometry and excitability. eLife 2020; 9:53432. [PMID: 32223890 PMCID: PMC7170651 DOI: 10.7554/elife.53432] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 03/30/2020] [Indexed: 12/29/2022] Open
Abstract
In most vertebrate neurons, action potentials are triggered at the distal end of the axon initial segment (AIS). Both position and length of the AIS vary across and within neuron types, with activity, development and pathology. What is the impact of AIS geometry on excitability? Direct empirical assessment has proven difficult because of the many potential confounding factors. Here, we carried a principled theoretical analysis to answer this question. We provide a simple formula relating AIS geometry and sodium conductance density to the somatic voltage threshold. A distal shift of the AIS normally produces a (modest) increase in excitability, but we explain how this pattern can reverse if a hyperpolarizing current is present at the AIS, due to resistive coupling with the soma. This work provides a theoretical tool to assess the significance of structural AIS plasticity for electrical function.
Collapse
Affiliation(s)
- Sarah Goethals
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Romain Brette
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| |
Collapse
|
15
|
Didonna A, Opal P. The role of neurofilament aggregation in neurodegeneration: lessons from rare inherited neurological disorders. Mol Neurodegener 2019; 14:19. [PMID: 31097008 PMCID: PMC6524292 DOI: 10.1186/s13024-019-0318-4] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 04/26/2019] [Indexed: 12/13/2022] Open
Abstract
Many neurodegenerative disorders, including Parkinson's, Alzheimer's, and amyotrophic lateral sclerosis, are well known to involve the accumulation of disease-specific proteins. Less well known are the accumulations of another set of proteins, neuronal intermediate filaments (NFs), which have been observed in these diseases for decades. NFs belong to the family of cytoskeletal intermediate filament proteins (IFs) that give cells their shape; they determine axonal caliber, which controls signal conduction; and they regulate the transport of synaptic vesicles and modulate synaptic plasticity by binding to neurotransmitter receptors. In the last two decades, a number of rare disorders caused by mutations in genes that encode NFs or regulate their metabolism have been discovered. These less prevalent disorders are providing novel insights into the role of NF aggregation in the more common neurological disorders.
Collapse
Affiliation(s)
- Alessandro Didonna
- Department of Neurology and Weill Institute for Neurosciences, University of California at San Francisco, San Francisco, CA, 94158, USA
| | - Puneet Opal
- Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA. .,Department of Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA.
| |
Collapse
|
16
|
Kim GW, Kim YH, Park K, Jeong GW. A comparative study of white matter volume between postoperative female-to-male transsexuals and healthy female. Int J Impot Res 2019; 31:432-438. [PMID: 30679768 DOI: 10.1038/s41443-019-0111-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 08/14/2018] [Accepted: 12/03/2018] [Indexed: 12/21/2022]
Abstract
Cross-sex hormones in female-to-male (FtM) transsexuals play a crucial role in brain plasticity. Morphological study associated with white matter (WM) volume in postoperative FtM transsexuals receiving cross-sex hormones has not been published yet. This study was performed to discriminate the regional WM volume differences between postoperative FtM transsexuals and female controls using voxel-based morphometry (VBM) and further to assess the correlations between regional volume variations and cross-sex hormones. WM volume was assessed in 12 postoperative FtM transsexuals receiving cross-sex hormones with 16 age-matched female controls. WM volume was processed using SPM8 software with diffeomorphic anatomical registration via an exponentiated Lie algebra (DARTEL) algorithm. Serum sex hormones, including estriol, free testosterone (free-T), estradiol, follicle-stimulating hormone, and luteinizing hormone were measured. Postoperative FtM transsexuals showed significantly (p < 0.05) larger WM volumes in the inferior parietal lobule, postcentral gyrus, and middle temporal gyrus compared with female controls. However, there were no brain areas with larger WM volume in female controls compared with FtM transsexuals. WM volumes of the inferior parietal lobule and middle temporal gyrus in FtM transsexuals were positively correlated with the levels of free-T. This study revealed WM volume change and its correlation with free-T level in postoperative FtM transsexuals. These findings will improve our understanding of the morphometric changes in FtM transsexuals under cross-sex hormone therapy.
Collapse
Affiliation(s)
- Gwang-Won Kim
- Advanced Institute of Aging Science, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Yun-Hyeon Kim
- Department of Radiology, Chonnam National University Hospital, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea
| | - Kwangsung Park
- Advanced Institute of Aging Science, Chonnam National University, Gwangju, 61186, Republic of Korea.,Department of Urology, Chonnam National University Hospital, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea
| | - Gwang-Woo Jeong
- Department of Radiology, Chonnam National University Hospital, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea.
| |
Collapse
|
17
|
Sun S, Babola T, Pregernig G, So KS, Nguyen M, Su SSM, Palermo AT, Bergles DE, Burns JC, Müller U. Hair Cell Mechanotransduction Regulates Spontaneous Activity and Spiral Ganglion Subtype Specification in the Auditory System. Cell 2018; 174:1247-1263.e15. [PMID: 30078710 PMCID: PMC6429032 DOI: 10.1016/j.cell.2018.07.008] [Citation(s) in RCA: 220] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 04/23/2018] [Accepted: 07/02/2018] [Indexed: 01/06/2023]
Abstract
Type I spiral ganglion neurons (SGNs) transmit sound information from cochlear hair cells to the CNS. Using transcriptome analysis of thousands of single neurons, we demonstrate that murine type I SGNs consist of subclasses that are defined by the expression of subsets of transcription factors, cell adhesion molecules, ion channels, and neurotransmitter receptors. Subtype specification is initiated prior to the onset of hearing during the time period when auditory circuits mature. Gene mutations linked to deafness that disrupt hair cell mechanotransduction or glutamatergic signaling perturb the firing behavior of SGNs prior to hearing onset and disrupt SGN subtype specification. We thus conclude that an intact hair cell mechanotransduction machinery is critical during the pre-hearing period to regulate the firing behavior of SGNs and their segregation into subtypes. Because deafness is frequently caused by defects in hair cells, our findings have significant ramifications for the etiology of hearing loss and its treatment.
Collapse
Affiliation(s)
- Shuohao Sun
- The Solomon Snyder Department of Neuroscience and Department of Otolaryngology, Head and Neck Surgery, Johns Hopkins University School of Medicine, 725 N. Wolfe Street, Baltimore, MD 21205, USA
| | - Travis Babola
- The Solomon Snyder Department of Neuroscience and Department of Otolaryngology, Head and Neck Surgery, Johns Hopkins University School of Medicine, 725 N. Wolfe Street, Baltimore, MD 21205, USA
| | - Gabriela Pregernig
- Decibel Therapeutics, 1325 Boylston Street, Suite 500, Boston, MA 02215, USA
| | - Kathy S So
- Decibel Therapeutics, 1325 Boylston Street, Suite 500, Boston, MA 02215, USA
| | - Matthew Nguyen
- Decibel Therapeutics, 1325 Boylston Street, Suite 500, Boston, MA 02215, USA
| | - Shin-San M Su
- Decibel Therapeutics, 1325 Boylston Street, Suite 500, Boston, MA 02215, USA
| | - Adam T Palermo
- Decibel Therapeutics, 1325 Boylston Street, Suite 500, Boston, MA 02215, USA
| | - Dwight E Bergles
- The Solomon Snyder Department of Neuroscience and Department of Otolaryngology, Head and Neck Surgery, Johns Hopkins University School of Medicine, 725 N. Wolfe Street, Baltimore, MD 21205, USA
| | - Joseph C Burns
- Decibel Therapeutics, 1325 Boylston Street, Suite 500, Boston, MA 02215, USA.
| | - Ulrich Müller
- The Solomon Snyder Department of Neuroscience and Department of Otolaryngology, Head and Neck Surgery, Johns Hopkins University School of Medicine, 725 N. Wolfe Street, Baltimore, MD 21205, USA.
| |
Collapse
|
18
|
Neuronal Subtype Determines Herpes Simplex Virus 1 Latency-Associated-Transcript Promoter Activity during Latency. J Virol 2018; 92:JVI.00430-18. [PMID: 29643250 DOI: 10.1128/jvi.00430-18] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 04/06/2018] [Indexed: 12/16/2022] Open
Abstract
Herpes simplex virus (HSV) latency in neurons remains poorly understood, and the heterogeneity of the sensory nervous system complicates mechanistic studies. In this study, we used primary culture of adult trigeminal ganglion (TG) mouse neurons in microfluidic devices and an in vivo model to examine the subtypes of sensory neurons involved in HSV latency. HSV-infected neurofilament heavy-positive (NefH+) neurons were more likely to express latency-associated transcripts (LATs) than infected neurofilament heavy-negative (NefH-) neurons. This differential expression of the LAT promoter correlated with differences in HSV-1 early infection that manifested as differences in the efficiency with which HSV particles reached the cell body following infection at the distal axon. In vivo, we further identified a specific subset of NefH+ neurons which coexpressed calcitonin gene-related peptide α (NefH+ CGRP+ neurons) as the sensory neuron subpopulation with the highest LAT promoter activity following HSV-1 infection. Finally, an early-phase reactivation assay showed HSV-1 reactivating in NefH+ CGRP+ neurons, although other sensory neuron subpopulations were also involved. Together, these results show that sensory neurons expressing neurofilaments exhibit enhanced LAT promoter activity. We hypothesize that the reduced efficiency of HSV-1 invasion at an early phase of infection may promote efficient establishment of latency in NefH+ neurons due to initiation of the antiviral state preceding arrival of the virus at the neuronal cell body. While the outcome of HSV-1 infection of neurons is determined by a broad variety of factors in vivo, neuronal subtypes are likely to play differential roles in modulating the establishment of latent infection.IMPORTANCE Two pivotal properties of HSV-1 make it a successful pathogen. First, it infects neurons, which are immune privileged. Second, it establishes latency in these neurons. Together, these properties allow HSV to persist for the lifetime of its host. Neurons are diverse and highly organized cells, with specific anatomical, physiological, and molecular characteristics. Previous work has shown that establishment of latency by HSV-1 does not occur equally in all types of neurons. Our results show that the kinetics of HSV infection and the levels of latency-related gene expression differ in certain types of neurons. The neuronal subtype infected by HSV is therefore a critical determinant of the outcome of infection and latency.
Collapse
|
19
|
Cerebrospinal Fluid from Patients with Sporadic Amyotrophic Lateral Sclerosis Induces Degeneration of Motor Neurons Derived from Human Embryonic Stem Cells. Mol Neurobiol 2018; 56:1014-1034. [PMID: 29858777 DOI: 10.1007/s12035-018-1149-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 05/23/2018] [Indexed: 12/15/2022]
Abstract
Disease modeling has become challenging in the context of amyotrophic lateral sclerosis (ALS), as obtaining viable spinal motor neurons from postmortem patient tissue is an unlikely possibility. Limitations in the animal models due to their phylogenetic distance from human species hamper the success of translating possible findings into therapeutic options. Accordingly, there is a need for developing humanized models as a lead towards identifying successful therapeutic possibilities. In this study, human embryonic stem cells-BJNHem20-were differentiated into motor neurons expressing HB9, Islet1, and choline acetyl transferase using retinoic acid and purmorphamine. These motor neurons discharged spontaneous action potentials with two different frequencies (< 5 and > 5 Hz), and majority of them were principal neurons firing with < 5 Hz. Exposure to cerebrospinal fluid from ALS patients for 48 h induced several degenerative changes in the motor neurons as follows: cytoplasmic changes such as beading of neurites and vacuolation; morphological alterations, viz., dilation and vacuolation of mitochondria, curled and closed Golgi architecture, dilated endoplasmic reticulum, and chromatin condensation in the nucleus; lowered activity of different mitochondrial complex enzymes; reduced expression of brain-derived neurotrophic factor; up-regulated neurofilament phosphorylation and hyperexcitability represented by increased number of spikes. All these changes along with the enhanced expression of pro-apoptotic proteins-Bax and caspase 9-culminated in the death of motor neurons.
Collapse
|
20
|
Overexpression of MAP2 and NF-H Associated with Dendritic Pathology in the Spinal Cord of Mice Infected with Rabies Virus. Viruses 2018; 10:v10030112. [PMID: 29509660 PMCID: PMC5869505 DOI: 10.3390/v10030112] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Revised: 01/29/2018] [Accepted: 02/08/2018] [Indexed: 12/13/2022] Open
Abstract
Rabies is a viral infection that targets the nervous system, specifically neurons. The clinical manifestations of the disease are dramatic and their outcome fatal; paradoxically, conventional histopathological descriptions reveal only subtle changes in the affected nervous tissue. Some researchers have considered that the pathophysiology of rabies is based more on biochemical changes than on structural alterations, as is the case with some psychiatric diseases. However, we believe that it has been necessary to resort to other methods that allow us to analyze the effect of the infection on neurons. The Golgi technique is the gold standard for studying the morphology of all the components of a neuron and the cytoskeletal proteins are the structural support of dendrites and axons. We have previously shown, in the mouse cerebral cortex and now with this work in spinal cord, that rabies virus generates remarkable alterations in the morphological pattern of the neurons and that this effect is associated with the increase in the expression of two cytoskeletal proteins (MAP2 and NF-H). It is necessary to deepen the investigation of the pathogenesis of rabies in order to find therapeutic alternatives to a disease to which the World Health Organization classifies as a neglected disease.
Collapse
|
21
|
Gatto RG, Li W, Magin RL. Diffusion tensor imaging identifies presymptomatic axonal degeneration in the spinal cord of ALS mice. Brain Res 2017; 1679:45-52. [PMID: 29175489 DOI: 10.1016/j.brainres.2017.11.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 11/14/2017] [Accepted: 11/18/2017] [Indexed: 12/11/2022]
Abstract
Extensive pathological evidence indicates that axonal degeneration represents an early and critical event in amyotrophic lateral sclerosis (ALS). Unfortunately, few MRI studies have focused in the early detection of white matter (WM) alterations in the spinal cord region. To unveil these WM changes, we performed high resolution diffusion tensor imaging (DTI) and correlated the results with histological analysis of adjacent slices taken from the spinal cords of presymptomatic mice. The DTI studies demonstrated a significant reduction in fractional anisotropy (FA) as well as axial diffusivities (AD) and an increase in radial diffusivity (RD), predominantly at lower segments of the spinal cord. Increases in FA and a reduction in AD and RD were observed in spinal cord (SC) gray matter (GM). Diffusion changes are associated with early and progressive alterations in axonal connectivity following a distal to proximal progression. Histological data tagging neuronal, axonal and glial cell markers demonstrated presymptomatic alterations in spinal cord WM and GM. This study demonstrates that DTI methods are optimal preclinical imaging tools to detect structural anomalies in WM and GM spinal cord during early stages of the disease.
Collapse
Affiliation(s)
- Rodolfo G Gatto
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, College of Medicine, Chicago, IL 60612, USA.
| | - Weiguo Li
- Department of Bioengineering, University of Illinois at Chicago, School of Engineering, Chicago, IL 60612, USA
| | - Richard L Magin
- Department of Bioengineering, University of Illinois at Chicago, School of Engineering, Chicago, IL 60612, USA
| |
Collapse
|
22
|
Zhao J, Brown K, Liem RKH. Abnormal neurofilament inclusions and segregations in dorsal root ganglia of a Charcot-Marie-Tooth type 2E mouse model. PLoS One 2017; 12:e0180038. [PMID: 28654681 PMCID: PMC5487060 DOI: 10.1371/journal.pone.0180038] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 06/08/2017] [Indexed: 11/20/2022] Open
Abstract
Charcot-Marie-Tooth (CMT) disease or hereditary motor and sensory neuropathy is the most prevalent inherited peripheral neuropathy and is associated with over 90 causative genes. Mutations in neurofilament light polypeptide gene, NEFL cause CMT2E, an axonal form of CMT that results in abnormal structures and/or functions of peripheral axons in spinal cord motor neurons and dorsal root ganglion neurons. We have previously generated and characterized a knock-in mouse model of CMT2E with the N98S mutation in Nefl that presented with multiple inclusions in spinal cord neurons. In this report, we conduct immunofluorescence studies of cultured dorsal root ganglia (DRG) from NeflN98S/+ mice, and show that inclusions found in DRG neurites can occur in embryonic stages. Ultrastructural analyses reveal that the inclusions are disordered neurofilaments packed in high density, segregated from other organelles. Immunochemical studies show decreased NFL protein levels in DRG, cerebellum and spinal cord in NeflN98S/+ mice, and total NFL protein pool is shifted toward the triton-insoluble fraction. Our findings reveal the nature of the inclusions in NeflN98S/+ mice, provide useful information to understand mechanisms of CMT2E disease, and identify DRG from NeflN98S/+ mice as a useful cell line model for therapeutic discoveries.
Collapse
Affiliation(s)
- Jian Zhao
- Department of Pathology and Cell Biology, Columbia University College of Physicians & Surgeons, New York, New York, United States of America
| | - Kristy Brown
- Department of Pathology and Cell Biology, Columbia University College of Physicians & Surgeons, New York, New York, United States of America
| | - Ronald K. H. Liem
- Department of Pathology and Cell Biology and Taub Institute for Research in Alzheimer’s Disease and the Aging Brain, Columbia University College of Physicians & Surgeons, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
23
|
Shelukhina I, Mikhailov N, Abushik P, Nurullin L, Nikolsky EE, Giniatullin R. Cholinergic Nociceptive Mechanisms in Rat Meninges and Trigeminal Ganglia: Potential Implications for Migraine Pain. Front Neurol 2017; 8:163. [PMID: 28496430 PMCID: PMC5406407 DOI: 10.3389/fneur.2017.00163] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 04/07/2017] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Parasympathetic innervation of meninges and ability of carbachol, acetylcholine (ACh) receptor (AChR) agonist, to induce headaches suggests contribution of cholinergic mechanisms to primary headaches. However, neurochemical mechanisms of cholinergic regulation of peripheral nociception in meninges, origin place for headache, are almost unknown. METHODS Using electrophysiology, calcium imaging, immunohistochemistry, and staining of meningeal mast cells, we studied effects of cholinergic agents on peripheral nociception in rat hemiskulls and isolated trigeminal neurons. RESULTS Both ACh and carbachol significantly increased nociceptive firing in peripheral terminals of meningeal trigeminal nerves recorded by local suction electrode. Strong nociceptive firing was also induced by nicotine, implying essential role of nicotinic AChRs in control of excitability of trigeminal nerve endings. Nociceptive firing induced by carbachol was reduced by muscarinic antagonist atropine, whereas the action of nicotine was prevented by the nicotinic blocker d-tubocurarine but was insensitive to the TRPA1 antagonist HC-300033. Carbachol but not nicotine induced massive degranulation of meningeal mast cells known to release multiple pro-nociceptive mediators. Enzymes terminating ACh action, acetylcholinesterase (AChE) and butyrylcholinesterase, were revealed in perivascular meningeal nerves. The inhibitor of AChE neostigmine did not change the firing per se but induced nociceptive activity, sensitive to d-tubocurarine, after pretreatment of meninges with the migraine mediator CGRP. This observation suggested the pro-nociceptive action of endogenous ACh in meninges. Both nicotine and carbachol induced intracellular Ca2+ transients in trigeminal neurons partially overlapping with expression of capsaicin-sensitive TRPV1 receptors. CONCLUSION Trigeminal nerve terminals in meninges, as well as dural mast cells and trigeminal ganglion neurons express a repertoire of pro-nociceptive nicotinic and muscarinic AChRs, which could be activated by the ACh released from parasympathetic nerves. These receptors represent a potential target for novel therapeutic interventions in trigeminal pain and probably in migraine.
Collapse
Affiliation(s)
- Irina Shelukhina
- Department of Neurobiology, A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland.,Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russian Federation
| | - Nikita Mikhailov
- Department of Neurobiology, A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Polina Abushik
- Department of Neurobiology, A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Leniz Nurullin
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, Kazan, Russian Federation.,Open Laboratory of Neuropharmacology, Kazan Federal University, Kazan, Russian Federation
| | - Evgeny E Nikolsky
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, Kazan, Russian Federation.,Open Laboratory of Neuropharmacology, Kazan Federal University, Kazan, Russian Federation
| | - Rashid Giniatullin
- Department of Neurobiology, A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland.,Laboratory of Neurobiology, Kazan Federal University, Kazan, Russian Federation
| |
Collapse
|
24
|
Yuan A, Rao MV, Veeranna, Nixon RA. Neurofilaments and Neurofilament Proteins in Health and Disease. Cold Spring Harb Perspect Biol 2017; 9:9/4/a018309. [PMID: 28373358 DOI: 10.1101/cshperspect.a018309] [Citation(s) in RCA: 495] [Impact Index Per Article: 61.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
SUMMARYNeurofilaments (NFs) are unique among tissue-specific classes of intermediate filaments (IFs) in being heteropolymers composed of four subunits (NF-L [neurofilament light]; NF-M [neurofilament middle]; NF-H [neurofilament heavy]; and α-internexin or peripherin), each having different domain structures and functions. Here, we review how NFs provide structural support for the highly asymmetric geometries of neurons and, especially, for the marked radial expansion of myelinated axons crucial for effective nerve conduction velocity. NFs in axons extensively cross-bridge and interconnect with other non-IF components of the cytoskeleton, including microtubules, actin filaments, and other fibrous cytoskeletal elements, to establish a regionally specialized network that undergoes exceptionally slow local turnover and serves as a docking platform to organize other organelles and proteins. We also discuss how a small pool of oligomeric and short filamentous precursors in the slow phase of axonal transport maintains this network. A complex pattern of phosphorylation and dephosphorylation events on each subunit modulates filament assembly, turnover, and organization within the axonal cytoskeleton. Multiple factors, and especially turnover rate, determine the size of the network, which can vary substantially along the axon. NF gene mutations cause several neuroaxonal disorders characterized by disrupted subunit assembly and NF aggregation. Additional NF alterations are associated with varied neuropsychiatric disorders. New evidence that subunits of NFs exist within postsynaptic terminal boutons and influence neurotransmission suggests how NF proteins might contribute to normal synaptic function and neuropsychiatric disease states.
Collapse
Affiliation(s)
- Aidong Yuan
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, New York 10962.,Department of Psychiatry, New York University School of Medicine, New York, New York 10016
| | - Mala V Rao
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, New York 10962.,Department of Psychiatry, New York University School of Medicine, New York, New York 10016
| | - Veeranna
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, New York 10962.,Department of Psychiatry, New York University School of Medicine, New York, New York 10016
| | - Ralph A Nixon
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, New York 10962.,Department of Psychiatry, New York University School of Medicine, New York, New York 10016.,Cell Biology, New York University School of Medicine, New York, New York 10016
| |
Collapse
|
25
|
|
26
|
Li Y, Zhang L, Kallakuri S, Cohen A, Cavanaugh JM. Correlation of mechanical impact responses and biomarker levels: A new model for biomarker evaluation in TBI. J Neurol Sci 2015; 359:280-6. [PMID: 26671128 DOI: 10.1016/j.jns.2015.08.035] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 08/19/2015] [Accepted: 08/20/2015] [Indexed: 12/19/2022]
Abstract
A modified Marmarou impact acceleration model was used to help screen biomarkers to assess brain injury severity. Anesthetized male Sprague-Dawley rats were subjected to a closed head injury from 1.25, 1.75 and 2.25 m drop heights. Linear and angular responses of the head were measured in vivo. 24h after impact, cerebrospinal fluid (CSF) and serum were collected. CSF and serum levels of phosphorylated neurofilament heavy (pNF-H), glial fibrillary acidic protein (GFAP), interleukin 6 (IL-6), and amyloid beta (Aβ) 1-42 were assessed by enzyme-linked immunosorbent assay (ELISA). Compared to controls, significantly higher CSF and serum pNF-H levels were observed in all impact groups, except between 1.25 m and control in serum. Furthermore, CSF and serum pNF-H levels were significantly different between the impact groups. For GFAP, both CSF and serum levels were significantly higher at 2.25 m compared to 1.75 m, 1.25 m and controls. There was no significant difference in CSF and serum GFAP levels between 1.75 m and 1.25 m, although both groups were significantly higher than control. TBI rats also showed significantly higher levels of IL-6 versus control in both CSF and serum, but no significant difference was observed between each impact group. Levels of Aβ were not significantly different between groups. Pearson's correlation analysis showed pNF-H and GFAP levels in CSF and serum had positive correlation with power (rate of impact energy), followed by average linear acceleration and surface righting (p<0.01), which were good predictors for traumatic axonal injury according to histologic assessment in our previous study, suggesting that they are directly related to the injury mechanism. The model used in this study showed a unique ability in elucidating the relationship between biomarker levels and severity of the mechanical trauma to the brain.
Collapse
Affiliation(s)
- Yan Li
- Department of Biomedical Engineering, Wayne State University, 818 W. Hancock Street, Detroit, MI 48201, United States
| | - Liying Zhang
- Department of Biomedical Engineering, Wayne State University, 818 W. Hancock Street, Detroit, MI 48201, United States.
| | - Srinivasu Kallakuri
- Department of Biomedical Engineering, Wayne State University, 818 W. Hancock Street, Detroit, MI 48201, United States
| | - Abigail Cohen
- Department of Biomedical Engineering, Wayne State University, 818 W. Hancock Street, Detroit, MI 48201, United States
| | - John M Cavanaugh
- Department of Biomedical Engineering, Wayne State University, 818 W. Hancock Street, Detroit, MI 48201, United States
| |
Collapse
|
27
|
Yuan A, Hassinger L, Rao MV, Julien JP, Miller CCJ, Nixon RA. Dissociation of Axonal Neurofilament Content from Its Transport Rate. PLoS One 2015. [PMID: 26208164 PMCID: PMC4514674 DOI: 10.1371/journal.pone.0133848] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The axonal cytoskeleton of neurofilament (NF) is a long-lived network of fibrous elements believed to be a stationary structure maintained by a small pool of transported cytoskeletal precursors. Accordingly, it may be predicted that NF content in axons can vary independently from the transport rate of NF. In the present report, we confirm this prediction by showing that human NFH transgenic mice and transgenic mice expressing human NFL Ser55 (Asp) develop nearly identical abnormal patterns of NF accumulation and distribution in association with opposite changes in NF slow transport rates. We also show that the rate of NF transport in wild-type mice remains constant along a length of the optic axon where NF content varies 3-fold. Moreover, knockout mice lacking NFH develop even more extreme (6-fold) proximal to distal variation in NF number, which is associated with a normal wild-type rate of NF transport. The independence of regional NF content and NF transport is consistent with previous evidence suggesting that the rate of incorporation of transported NF precursors into a metabolically stable stationary cytoskeletal network is the major determinant of axonal NF content, enabling the generation of the striking local variations in NF number seen along axons.
Collapse
Affiliation(s)
- Aidong Yuan
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, New York, United States of America
- Department of Psychiatry, New York University School of Medicine, New York, New York, United States of America
- * E-mail: (AY); (RAN)
| | - Linda Hassinger
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, Massachusetts, United States of America
| | - Mala V. Rao
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, New York, United States of America
- Department of Psychiatry, New York University School of Medicine, New York, New York, United States of America
| | - Jean-Pierre Julien
- Centre de Recherche du Centre Hospitalier de l'Université Laval, Département d'anatomie et physiologie de l'Université Laval, Québec, Canada
| | - Christopher C. J. Miller
- Department of Neuroscience, Institute of Psychiatry, Kings College London, London, United Kingdom
- Clinical Neurosciences, Institute of Psychiatry, Kings College London, London, United Kingdom
| | - Ralph A. Nixon
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, New York, United States of America
- Department of Psychiatry, New York University School of Medicine, New York, New York, United States of America
- Department of Cell Biology, New York University School of Medicine, New York, New York, United States of America
- * E-mail: (AY); (RAN)
| |
Collapse
|
28
|
Broms J, Antolin-Fontes B, Tingström A, Ibañez-Tallon I. Conserved expression of the GPR151 receptor in habenular axonal projections of vertebrates. J Comp Neurol 2015; 523:359-80. [PMID: 25116430 PMCID: PMC4270839 DOI: 10.1002/cne.23664] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Revised: 08/06/2014] [Accepted: 08/11/2014] [Indexed: 12/11/2022]
Abstract
The habenula is a phylogenetically conserved brain structure in the epithalamus. It is a major node in the information flow between fronto-limbic brain regions and monoaminergic brainstem nuclei, and is thus anatomically and functionally ideally positioned to regulate emotional, motivational, and cognitive behaviors. Consequently, the habenula may be critically important in the pathophysiology of psychiatric disorders such as addiction and depression. Here we investigated the expression pattern of GPR151, a G protein-coupled receptor (GPCR), whose mRNA has been identified as highly and specifically enriched in habenular neurons by in situ hybridization and translating ribosome affinity purification (TRAP). In the present immunohistochemical study we demonstrate a pronounced and highly specific expression of the GPR151 protein in the medial and lateral habenula of rodent brain. Specific expression was also seen in efferent habenular fibers projecting to the interpeduncular nucleus, the rostromedial tegmental area, the rhabdoid nucleus, the mesencephalic raphe nuclei, and the dorsal tegmental nucleus. Using confocal microscopy and quantitative colocalization analysis, we found that GPR151-expressing axons and terminals overlap with cholinergic, substance P-ergic, and glutamatergic markers. Virtually identical expression patterns were observed in rat, mouse, and zebrafish brains. Our data demonstrate that GPR151 is highly conserved, specific for a subdivision of the habenular neurocircuitry, and constitutes a promising novel target for psychiatric drug development.
Collapse
Affiliation(s)
- Jonas Broms
- Psychiatric Neuromodulation Unit, Clinical Sciences, Lund University, Lund, Sweden
| | | | - Anders Tingström
- Psychiatric Neuromodulation Unit, Clinical Sciences, Lund University, Lund, Sweden
| | - Ines Ibañez-Tallon
- Laboratory of Molecular Biology, The Rockefeller University, New York, U.S.A
| |
Collapse
|
29
|
Neuroproteomics in the auditory brainstem: Candidate proteins for ultrafast and precise information processing. Mol Cell Neurosci 2015; 64:9-23. [DOI: 10.1016/j.mcn.2014.08.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Revised: 07/25/2014] [Accepted: 08/12/2014] [Indexed: 12/18/2022] Open
|
30
|
Kang MH, Law-Davis S, Balaratnasingam C, Yu DY. Sectoral variations in the distribution of axonal cytoskeleton proteins in the human optic nerve head. Exp Eye Res 2014; 128:141-50. [DOI: 10.1016/j.exer.2014.10.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 09/12/2014] [Accepted: 10/06/2014] [Indexed: 01/15/2023]
|
31
|
Gilbert MT, Soderstrom K. Developmental but not adult cannabinoid treatments persistently alter axonal and dendritic morphology within brain regions important for zebra finch vocal learning. Brain Res 2014; 1558:57-73. [PMID: 24594017 DOI: 10.1016/j.brainres.2014.02.039] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Revised: 02/21/2014] [Accepted: 02/22/2014] [Indexed: 01/29/2023]
Abstract
Prior work shows developmental cannabinoid exposure alters zebra finch vocal development in a manner associated with altered CNS physiology, including changes in patterns of CB1 receptor immunoreactivity, endocannabinoid concentrations and dendritic spine densities. These results raise questions about the selectivity of developmental cannabinoid effects: are they a consequence of a generalized developmental disruption, or are effects produced through more selective and distinct interactions with biochemical pathways that control receptor, endogenous ligand and dendritic spine dynamics? To begin to address this question we have examined effects of developmental cannabinoid exposure on the pattern and density of expression of proteins critical to dendritic (MAP2) and axonal (Nf-200) structure to determine the extent to which dendritic vs. axonal neuronal morphology may be altered. Results demonstrate developmental, but not adult cannabinoid treatments produce generalized changes in expression of both dendritic and axonal cytoskeletal proteins within brain regions and cells known to express CB1 cannabinoid receptors. Results clearly demonstrate that cannabinoid exposure during a period of sensorimotor development, but not adulthood, produce profound effects upon both dendritic and axonal morphology that persist through at least early adulthood. These findings suggest an ability of exogenous cannabinoids to alter general processes responsible for normal brain development. Results also further implicate the importance of endocannabinoid signaling to peri-pubertal periods of adolescence, and underscore potential consequences of cannabinoid abuse during periods of late-postnatal CNS development.
Collapse
Affiliation(s)
- Marcoita T Gilbert
- Department of Pharmacology and Toxicology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, United States
| | - Ken Soderstrom
- Department of Pharmacology and Toxicology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, United States.
| |
Collapse
|
32
|
Paus T, Pesaresi M, French L. White matter as a transport system. Neuroscience 2014; 276:117-25. [PMID: 24508743 DOI: 10.1016/j.neuroscience.2014.01.055] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Revised: 01/14/2014] [Accepted: 01/29/2014] [Indexed: 12/14/2022]
Abstract
There are two ways to picture white matter: as a grid of electrical wires or a network of roads. The first metaphor captures the classical function of an axon as conductor of action potentials (and information) from one brain region to another. The second one points to the important role of axons in a bi-directional transport of biological molecules and organelles between the cell body and synapse. Given the wide variety of such cargoes, a well-functioning axonal transport is critical for a number of processes, including neurotransmission, metabolism and viability of neurons. This selective review will emphasize the need for considering axonal transport when interpreting functional consequences of inter-individual variations in the structural properties of white matter. We start by describing the space occupied by white matter and techniques used in vivo for its characterization. We then provide examples of key features of maturation and aging of white matter, as well as some of the common abnormalities observed in neurodevelopmental and neurodegenerative disorders. Next, we review work that motivated our focus on axonal diameter, and explain the relationships between transport and cytoskeleton within the axon. We will conclude by describing molecular machinery of axonal transport and genes that may contribute to inter-individual variations in axonal diameter and axonal transport.
Collapse
Affiliation(s)
- T Paus
- Rotman Research Institute, University of Toronto, Toronto, Canada.
| | - M Pesaresi
- Rotman Research Institute, University of Toronto, Toronto, Canada
| | - L French
- Rotman Research Institute, University of Toronto, Toronto, Canada
| |
Collapse
|
33
|
Edwards AVG, Edwards GJ, Schwämmle V, Saxtorph H, Larsen MR. Spatial and Temporal Effects in Protein Post-translational Modification Distributions in the Developing Mouse Brain. J Proteome Res 2013; 13:260-7. [DOI: 10.1021/pr4002977] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Alistair V. G. Edwards
- Department of Biochemistry
and Molecular Biology, University of Southern Denmark, Campusvej 55, Odense, DK 5230, Denmark
| | | | - Veit Schwämmle
- Department of Biochemistry
and Molecular Biology, University of Southern Denmark, Campusvej 55, Odense, DK 5230, Denmark
| | - Henrik Saxtorph
- Biomedical Laboratory, Odense University Hospital, Winsløwparken 23, Odense, DK 5000, Denmark
| | - Martin R. Larsen
- Department of Biochemistry
and Molecular Biology, University of Southern Denmark, Campusvej 55, Odense, DK 5230, Denmark
| |
Collapse
|
34
|
Craiu D. What is special about the adolescent (JME) brain? Epilepsy Behav 2013; 28 Suppl 1:S45-51. [PMID: 23756479 DOI: 10.1016/j.yebeh.2012.12.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Accepted: 12/09/2012] [Indexed: 01/23/2023]
Abstract
Juvenile myoclonic epilepsy (JME) involves cortico-thalamo-cortical networks. Thalamic, frontal gray matter, connectivity, and neurotransmitter disturbances have been demonstrated by structural/functional imaging studies. Few patients with JME show mutations in genes coding ion channels or GABAA (gamma-aminobutyric acid) receptor subunits. Recent research points to EFHC1 gene mutations leading to microdysgenesis and possible aberrant circuitry. Imaging studies have shown massive structural/functional changes of normally developing adolescent brain structures maturing at strikingly different rates and times. Gray matter (GM) volume diminishes in cortical areas (frontal and parietal) and deep structures (anterior thalamus, putamen, and caudate). Diffusion tensor imaging (DTI) findings support continued microstructural change in WM (white matter) during late adolescence with robust developmental changes in thalamocortical connectivity. The GABAA receptor distribution and specific receptor subunits' expression patterns change with age from neonate to adolescent/adult, contributing to age-related changes in brain excitability. Hormonal influence on brain structure development during adolescence is presented. Possible implications of brain changes during adolescence on the course of JME are discussed.
Collapse
Affiliation(s)
- Dana Craiu
- Pediatric Neurology Clinic, Alexandru Obregia Clinical Hospital, Carol Davila University of Medicine, Bucharest, Romania.
| |
Collapse
|
35
|
|
36
|
Holmgren A, Bouhy D, Timmerman V. Neurofilament phosphorylation and their proline-directed kinases in health and disease. J Peripher Nerv Syst 2012; 17:365-76. [DOI: 10.1111/j.1529-8027.2012.00434.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
37
|
Rao MV, Yuan A, Campbell J, Kumar A, Nixon RA. The C-terminal domains of NF-H and NF-M subunits maintain axonal neurofilament content by blocking turnover of the stationary neurofilament network. PLoS One 2012; 7:e44320. [PMID: 23028520 PMCID: PMC3448626 DOI: 10.1371/journal.pone.0044320] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Accepted: 08/01/2012] [Indexed: 02/03/2023] Open
Abstract
Newly synthesized neurofilaments or protofilaments are incorporated into a highly stable stationary cytoskeleton network as they are transported along axons. Although the heavily phosphorylated carboxyl-terminal tail domains of the heavy and medium neurofilament (NF) subunits have been proposed to contribute to this process and particularly to stability of this structure, their function is still obscure. Here we show in NF-H/M tail deletion [NF-(H/M)(tailΔ)] mice that the deletion of both of these domains selectively lowers NF levels 3-6 fold along optic axons without altering either rates of subunit synthesis or the rate of slow axonal transport of NF. Pulse labeling studies carried out over 90 days revealed a significantly faster rate of disappearance of NF from the stationary NF network of optic axons in NF-(H/M)(tailΔ) mice. Faster NF disappearance was accompanied by elevated levels of NF-L proteolytic fragments in NF-(H/M)(tailΔ) axons. We conclude that NF-H and NF-M C-terminal domains do not normally regulate NF transport rates as previously proposed, but instead increase the proteolytic resistance of NF, thereby stabilizing the stationary neurofilament cytoskeleton along axons.
Collapse
Affiliation(s)
- Mala V Rao
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, New York, United States of America.
| | | | | | | | | |
Collapse
|
38
|
Lee WC, Kan D, Chen YY, Han SK, Lu KS, Chien CL. Suppression of extensive neurofilament phosphorylation rescues α-Internexin/peripherin-overexpressing PC12 cells from neuronal cell death. PLoS One 2012; 7:e43883. [PMID: 22952800 PMCID: PMC3428284 DOI: 10.1371/journal.pone.0043883] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2012] [Accepted: 07/26/2012] [Indexed: 11/23/2022] Open
Abstract
Intermediate filament (IF) overproduction induces abnormal accumulation of neuronal IF, which is a pathological indicator of some neurodegenerative disorders. In our study, α-Internexin- and peripherin-overexpressing PC12 cells (pINT-EGFP and pEGFP-peripherin) were used as models to study neuropathological pathways responsible for neurodegenerative diseases. Microarray data revealed that Cdk5-related genes were downregulated and Cdk5 regulatory subunit-associated protein 3 (GSK-3α and GSK-3β) were upregulated in pINT-EGFP cells. Increased expression of phosphorylated neurofilament and aberrant activation of Cdk5 and GSK-3β were detected in both pEGFP-peripherin and pINT-EGFP cells by Western blotting. In addition, pharmacological approaches to retaining viability of pINT-EGFP and pEGFP-peripherin cells were examined. Treatment with Cdk5 inhibitor and GSK-3β inhibitor significantly suppressed neuronal death. Dynamic changes of disaggregation of EGFP-peripherin and decrease in green fluorescence intensity were observed in pEGFP-peripherin and pINT-EGFP cells by confocal microscopy after GSK-3β inhibitor treatment. We conclude that inhibition of Cdk5 and GSK-3β suppresses neurofilament phosphorylation, slows down the accumulation of neuronal IF in the cytoplasm, and subsequently avoids damages to cell organelles. The results suggest that suppression of extensive neurofilament phosphorylation may be a potential strategy for ameliorating neuron death. The suppression of hyperphosphorylation of neuronal cytoskeletons with kinase inhibitors could be one of potential therapeutic treatments for neurodegenerative diseases.
Collapse
Affiliation(s)
- Wen-Ching Lee
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan, Republic of China
| | - Daphne Kan
- Center of Genomic Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan, Republic of China
| | - Yun-Yu Chen
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan, Republic of China
| | - Shan-Kuo Han
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan, Republic of China
| | - Kuo-Shyan Lu
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan, Republic of China
| | - Chung-Liang Chien
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan, Republic of China
- Center of Genomic Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan, Republic of China
- * E-mail:
| |
Collapse
|
39
|
Lee WC, Chen YY, Kan D, Chien CL. A neuronal death model: overexpression of neuronal intermediate filament protein peripherin in PC12 cells. J Biomed Sci 2012; 19:8. [PMID: 22252275 PMCID: PMC3282651 DOI: 10.1186/1423-0127-19-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2011] [Accepted: 01/17/2012] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Abnormal accumulation of neuronal intermediate filament (IF) is a pathological indicator of some neurodegenerative disorders. However, the underlying neuropathological mechanisms of neuronal IF accumulation remain unclear. A stable clone established from PC12 cells overexpressing a GFP-Peripherin fusion protein (pEGFP-Peripherin) was constructed for determining the pathway involved in neurodegeneration by biochemical, cell biology, and electronic microscopy approaches. In addition, pharmacological approaches to preventing neuronal death were also examined. RESULTS Results of this study showed that TUNEL positive reaction could be detected in pEGFP-Peripherin cells. Swollen mitochondria and endoplasmic reticulum (ER) were seen by electron microscopy in pEGFP-Peripherin cells on day 8 of nerve growth factor (NGF) treatment. Peripherin overexpression not only led to the formation of neuronal IF aggregate but also causes aberrant neuronal IF phosphorylation and mislocation. Western blots showed that calpain, caspase-12, caspase-9, and caspase-3 activity was upregulated. Furthermore, treatment with calpain inhibitor significantly inhibited cell death. CONCLUSIONS These results suggested that the cytoplasmic neuronal IF aggregate caused by peripherin overexpression may induce aberrant neuronal IF phosphorylation and mislocation subsequently trapped and indirectly damaged mitochondria and ER. We suggested that the activation of calpain, caspase-12, caspase-9, and caspase-3 were correlated to the dysfunction of the ER and mitochondria in our pEGFP-Peripherin cell model. The present study suggested that pEGFP-Peripherin cell clones could be a neuronal death model for future studies in neuronal IFs aggregate associated neurodegeneration.
Collapse
Affiliation(s)
- Wen-Ching Lee
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Jen-Ai Road, Taipei, 100, Taiwan
| | - Yun-Yu Chen
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Jen-Ai Road, Taipei, 100, Taiwan
| | - Daphne Kan
- Center of Genomic Medicine, National Taiwan University, Jen-Ai Road, Taipei, 100, Taiwan
| | - Chung-Liang Chien
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Jen-Ai Road, Taipei, 100, Taiwan
- Center of Genomic Medicine, National Taiwan University, Jen-Ai Road, Taipei, 100, Taiwan
| |
Collapse
|
40
|
Erratum to: Rostrocaudal Dynamics of CSF Biomarkers. Neurochem Res 2012. [DOI: 10.1007/s11064-011-0609-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
41
|
Lenz B, Müller CP, Stoessel C, Sperling W, Biermann T, Hillemacher T, Bleich S, Kornhuber J. Sex hormone activity in alcohol addiction: integrating organizational and activational effects. Prog Neurobiol 2011; 96:136-63. [PMID: 22115850 DOI: 10.1016/j.pneurobio.2011.11.001] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2011] [Revised: 11/03/2011] [Accepted: 11/08/2011] [Indexed: 01/06/2023]
Abstract
There are well-known sex differences in the epidemiology and etiopathology of alcohol dependence. Male gender is a crucial risk factor for the onset of alcohol addiction. A directly modifying role of testosterone in alcohol addiction-related behavior is well established. Sex hormones exert both permanent (organizational) and transient (activational) effects on the human brain. The sensitive period for these effects lasts throughout life. In this article, we present a novel early sex hormone activity model of alcohol addiction. We propose that early exposure to sex hormones triggers structural (organizational) neuroadaptations. These neuroadaptations affect cellular and behavioral responses to adult sex hormones, sensitize the brain's reward system to the reinforcing properties of alcohol and modulate alcohol addictive behavior later in life. This review outlines clinical findings related to the early sex hormone activity model of alcohol addiction (handedness, the second-to-fourth-finger length ratio, and the androgen receptor and aromatase) and includes clinical and preclinical literature regarding the activational effects of sex hormones in alcohol drinking behavior. Furthermore, we discuss the role of the hypothalamic-pituitary-adrenal and -gonadal axes and the opioid system in mediating the relationship between sex hormone activity and alcohol dependence. We conclude that a combination of exposure to sex hormones in utero and during early development contributes to the risk of alcohol addiction later in life. The early sex hormone activity model of alcohol addiction may prove to be a valuable tool in the development of preventive and therapeutic strategies.
Collapse
Affiliation(s)
- Bernd Lenz
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-University of Erlangen-Nuremberg, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Shea TB, Lee S. Neurofilament phosphorylation regulates axonal transport by an indirect mechanism: a merging of opposing hypotheses. Cytoskeleton (Hoboken) 2011; 68:589-95. [PMID: 21990272 DOI: 10.1002/cm.20535] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2011] [Accepted: 09/22/2011] [Indexed: 01/22/2023]
Abstract
Neurofilaments (NFs) are among the most abundant constituents of the axonal cytoskeleton. NFs consist of four subunits, termed NF-H, NF-M and NF-L, corresponding to heavy, medium and light in reference to their molecular mass and α-internexin. Phosphorylation of the C-terminal "sidearms" of NF-H and NF-M regulates the ability of NFs to form a cytoskeletal lattice that supports the mature axon. C-terminal phosphorylation events have classically been considered to regulate NF axonal transport. By contrast, studies demonstrating that NF axonal transport was not accelerated following sidearm deletion provided evidence that phosphorylation does not regulate NF transport. Herein, we demonstrate how comparison of transport and distribution of differentially phosphorylated NFs along axons identify common ground between these hypotheses and may resolve this controversy.
Collapse
Affiliation(s)
- Thomas B Shea
- Department of Biological Sciences, Center for Cellular Neurobiology and Neurodegeneration Research, University of Massachusetts-Lowell, Lowell, Massachusetts 01854, USA.
| | | |
Collapse
|
43
|
Cai J, Tuong CM, Zhang Y, Shields CB, Guo G, Fu H, Gozal D. Mouse intermittent hypoxia mimicking apnoea of prematurity: effects on myelinogenesis and axonal maturation. J Pathol 2011; 226:495-508. [PMID: 21953180 DOI: 10.1002/path.2980] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2011] [Revised: 07/24/2011] [Accepted: 08/04/2011] [Indexed: 01/09/2023]
Abstract
Premature babies are at high risk for both infantile apnoea and long-term neurobehavioural deficits. Recent studies suggest that diffuse structural changes in brain white matter are a positive predictor of poor cognitive outcomes. Since oligodendrocyte maturation, myelination, axon development, and synapse formation mainly occur in the third trimester of gestation and first postnatal year, infantile apnoea could lead to and/or exaggerate white matter impairments in preterm neonates. Therefore, we investigated oligodendroglia and axon development in a neonatal mouse model of intermittent hypoxia between postnatal days 2 and 10. During critical phases of central nervous system development, intermittent hypoxia induced hypomyelination in the corpus callosum, striatum, fornix, and cerebellum, but not in the pons or spinal cord. Intermittent hypoxia-elicited alterations in myelin-forming processes were reflected by decreased expression of myelin proteins, including MBP, PLP, MAG, and CNPase, possibly due to arrested maturation of oligodendrocytes. Ultrastructural abnormalities were apparent in the myelin sheath and axon. Immature oligodendrocytes were more vulnerable to neonatal intermittent hypoxia exposures than developing axons, suggesting that hypomyelination may contribute, at least partially, to axonal deficits. Insufficient neurofilament synthesis with anomalous components of neurofilament subunits, β-tubulin, and MAP2 isoforms indicated immaturity of axons in intermittent hypoxia-exposed mouse brains. In addition, down-regulation of synapsin I, synaptophysin, and Gap-43 phosphorylation suggested a potential stunt in axonogenesis and synaptogenesis. The region-selective and complex impairment in brain white matter induced by intermittent hypoxia was further associated with electrophysiological changes that may underlie long-term neurobehavioural sequelae.
Collapse
Affiliation(s)
- Jun Cai
- Kosair Children's Hospital Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY 40202, USA.
| | | | | | | | | | | | | |
Collapse
|
44
|
Blood and CSF Biomarker Dynamics in Multiple Sclerosis: Implications for Data Interpretation. Mult Scler Int 2011; 2011:823176. [PMID: 22096644 PMCID: PMC3195856 DOI: 10.1155/2011/823176] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2010] [Accepted: 02/16/2011] [Indexed: 01/09/2023] Open
Abstract
Background. Disability in multiple sclerosis (MS) is related to neuroaxonal degeneration. A reliable blood biomarker for neuroaxonal degeneration is needed. Objectives. To explore the relationship between cerebrospinal fluid (CSF) and serum concentrations of a protein biomarker for neuroaxonal degeneration, the neurofilaments heavy chain (NfH). Methods. An exploratory cross-sectional (n = 51) and longitudinal (n = 34) study on cerebrospinal fluid (CSF) and serum NfH phosphoform levels in patients with MS. The expanded disability status scale (EDSS), CSF, and serum levels of NfH-SMI34 and NfH-SMI35 were quantified at baseline. Disability progression was assessed at 3-year followup. Results. At baseline, patients with primary progressive MS (PPMS, EDSS 6) and secondary progressive MS (SPMS, EDSS 6) were more disabled compared to patients with relapsing remitting MS (RRMS, EDSS 2, P < .0001). Serum and CSF NfH phosphoform levels were not correlated. Baseline serum levels of the NfH-SMI34 were significantly (P < .05) higher in patients with PPMS (2.05 ng/mL) compared to SPMS (0.03 ng/mL) and RRMS (1.56 ng/mL). In SPMS higher serum than CSF NfH-SMI34 levels predicted disability progression from baseline (ΔEDSS 2, P < .05). In RRMS higher CSF than serum NfH-SMI35 levels predicted disability progression (ΔEDSS 2, P < .05). Conclusion. Serum and CSF NfH-SMI34 and NfH-SMI35 levels did not correlate with each other in MS. The quantitative relationship of CSF and serum NfH levels suggests that neuroaxonal degeneration of the central nervous system is the likely cause for disability progression in RRMS. In more severely disabled patients with PP/SPMS, subtle pathology of the peripheral nervous system cannot be excluded as an alternative source for blood NfH levels. Therefore, the interpretation of blood protein biomarker data in diseases of the central nervous system (CNS) should consider the possibility that pathology of the peripheral nervous system (PNS) may influence the results.
Collapse
|
45
|
Petzold A, Mondria T, Kuhle J, Rocca MA, Cornelissen J, te Boekhorst P, Lowenberg B, Giovannoni G, Filippi M, Kappos L, Hintzen R. Evidence for acute neurotoxicity after chemotherapy. Ann Neurol 2011; 68:806-15. [PMID: 21194151 DOI: 10.1002/ana.22169] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
OBJECTIVE Chronic neurotoxicity is a recognized long-term complication following chemotherapy in a range of diseases. Neurotoxicity adversely affects patients' quality of life. The objective of this study is to examine whether there is evidence of acute neurotoxicity. METHODS This prospective study included patients with secondary progressive multiple sclerosis (SPMS-BMT, n = 14) and hematological malignancies (HM-BMT, n = 17) receiving chemotherapy as preconditioning for bone marrow transplant. The control groups included SPMS patients matched for demographic and clinical data (SPMS-PL, n = 14) and healthy controls (n = 14). Neurodegeneration was assessed at baseline and longitudinally (months 1, 2, 3, 6, 9, 12, 24, and 36), combining a clinical scale for disability (Expanded Disability Status Scale [EDSS]), a serum protein biomarker for neurodegeneration (neurofilaments, NfH-SMI35), and brain atrophy measures (magnetic resonance imaging). RESULTS Disability progression was significantly more acute and severe following chemotherapy compared to placebo. Immediately after starting chemotherapy, serum NfH-SMI35 levels increased in 79% (p < 0.0001) of SPMS-BMT patients and 41% (p < 0.01) of HM-BMT patients compared to 0% of SPMS-PL patients or healthy controls. In SPMS-BMT serum NfH-SMI35 levels were > 100-fold higher 1 month after chemotherapy (29.73ng/ml) compared to baseline (0.28ng/ml, p < 0.0001). High serum NfH-SMI35 levels persisting for at least 3 months were associated with sustained disability progression on the EDSS (p < 0.05). Brain atrophy rates increased acutely in SPMS-BMT (-2.09) compared to SPMS-PL (-1.18, p < 0.05). INTERPRETATION Neurotoxicity is an unwanted acute side effect of aggressive chemotherapy.
Collapse
Affiliation(s)
- Axel Petzold
- Department of Neuroimmunology, UCL Institute of Neurology, Queen Square, London, United Kingdom.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Szaro BG, Strong MJ. Regulation of Cytoskeletal Composition in Neurons: Transcriptional and Post-transcriptional Control in Development, Regeneration, and Disease. ADVANCES IN NEUROBIOLOGY 2011. [DOI: 10.1007/978-1-4419-6787-9_24] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
|
47
|
Wang L, Brown A. A hereditary spastic paraplegia mutation in kinesin-1A/KIF5A disrupts neurofilament transport. Mol Neurodegener 2010; 5:52. [PMID: 21087519 PMCID: PMC3000839 DOI: 10.1186/1750-1326-5-52] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2010] [Accepted: 11/18/2010] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Hereditary spastic paraplegias are a group of neurological disorders characterized by progressive distal degeneration of the longest ascending and descending axons in the spinal cord, leading to lower limb spasticity and weakness. One of the dominantly inherited forms of this disease (spastic gait type 10, or SPG10) is caused by point mutations in kinesin-1A (also known as KIF5A), which is thought to be an anterograde motor for neurofilaments. RESULTS We investigated the effect of an SPG10 mutation in kinesin-1A (N256S-kinesin-1A) on neurofilament transport in cultured mouse cortical neurons using live-cell fluorescent imaging. N256S-kinesin-1A decreased both anterograde and retrograde neurofilament transport flux by decreasing the frequency of anterograde and retrograde movements. Anterograde velocity was not affected, whereas retrograde velocity actually increased. CONCLUSIONS These data reveal subtle complexities to the functional interdependence of the anterograde and retrograde neurofilament motors and they also raise the possibility that anterograde and retrograde neurofilament transport may be disrupted in patients with SPG10.
Collapse
Affiliation(s)
- Lina Wang
- Center for Molecular Neurobiology and Department of Neuroscience, The Ohio State University, Columbus, OH 43210, USA
| | - Anthony Brown
- Center for Molecular Neurobiology and Department of Neuroscience, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
48
|
Shimoshige Y, Enomoto R, Aoki T, Matsuoka N, Kaneko S. The involvement of aldose reductase in alterations to neurotrophin receptors and neuronal cytoskeletal protein mRNA levels in the dorsal root ganglion of streptozotocin-induced diabetic rats. Biol Pharm Bull 2010; 33:67-71. [PMID: 20045938 DOI: 10.1248/bpb.33.67] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Dorsal root ganglia (DRG) are recognized as one of the organs which are damaged in peripheral sensory diabetic neuropathy. In an experimental animal model, the alteration of the mRNA expression level of neurotrophins, their receptors and neuronal cytoskeletal protein have been reported. In this study, we examined whether these changes are improved by treatment with the aldose reductase inhibitor, zenarestat, in early-stage diabetic neuropathy of streptozotocin (STZ)-induced diabetic rats. Two weeks after the induction of diabetes mellitus by STZ treatment, zenarestat or a vehicle were given orally for two weeks. After the zenarestat treatment, the mRNA expression levels of neurotrophin receptors and neuronal cytoskeletal proteins in dorsal root ganglia were determined with a real-time polymerase chain reaction (PCR) method. Compared with the expression level of normal rats, a significant increase in Trk-C and Talpha1 alpha-tubulin and a decrease in neurofilament H mRNA expression level were observed in the DRG of STZ rats, while there were no significant changes in Trk-A, Trk-B, p75, neurofilament L, neurofilament M and betaIII tubulin mRNA expression. Zenarestat treatment significantly ameliorated the abnormal increase in Trk-C mRNA expression level. These data suggest that hyperactivation of the polyol pathway induces a deficit in neurotropism on peripheral sensory diabetic neuropathy.
Collapse
Affiliation(s)
- Yukinori Shimoshige
- Pharmacology Research Labs, Astellas Pharma Inc., Tsukuba, Ibaraki 305-8585, Japan.
| | | | | | | | | |
Collapse
|
49
|
Ilieva H, Polymenidou M, Cleveland DW. Non-cell autonomous toxicity in neurodegenerative disorders: ALS and beyond. ACTA ACUST UNITED AC 2010; 187:761-72. [PMID: 19951898 PMCID: PMC2806318 DOI: 10.1083/jcb.200908164] [Citation(s) in RCA: 809] [Impact Index Per Article: 53.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Selective degeneration and death of one or more classes of neurons is the defining feature of human neurodegenerative disease. Although traditionally viewed as diseases mainly affecting the most vulnerable neurons, in most instances of inherited disease the causative genes are widely—usually ubiquitously—expressed. Focusing on amyotrophic lateral sclerosis (ALS), especially disease caused by dominant mutations in Cu/Zn superoxide dismutase (SOD1), we review here the evidence that it is the convergence of damage developed within multiple cell types, including within neighboring nonneuronal supporting cells, which is crucial to neuronal dysfunction. Damage to a specific set of key partner cells as well as to vulnerable neurons may account for the selective susceptibility of neuronal subtypes in many human neurodegenerative diseases, including Huntington's disease (HD), Parkinson's disease (PD), prion disease, the spinal cerebellar ataxias (SCAs), and Alzheimer's disease (AD).
Collapse
Affiliation(s)
- Hristelina Ilieva
- Ludwig Institute for Cancer Research and Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | | | | |
Collapse
|
50
|
Hervé PY, Leonard G, Perron M, Pike B, Pitiot A, Richer L, Veillette S, Pausova Z, Paus T. Handedness, motor skills and maturation of the corticospinal tract in the adolescent brain. Hum Brain Mapp 2009; 30:3151-62. [PMID: 19235881 DOI: 10.1002/hbm.20734] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
With anatomical magnetic resonance imaging, the signal intensity of the corticospinal tract (CST) at the level of the internal capsule is often paradoxically similar to that of grey matter. As shown previously in histological studies, this is likely due to the presence of very large axons. We measured the apparent grey-matter density (aGMd) of the putative CST (pCST) in a large cohort of adolescents (n = 409, aged 12-18 years). We tested the following hypotheses: (1) The aGMd in the pCST shows a hemispheric asymmetry that is, in turn, related to hand preference; (2) the maturation of the CST during adolescence differs between both sexes, due to the influence of testosterone; (3) variations in aGMd in the pCST reflect inter-individual differences in manual skills. We confirmed the first two predictions. Thus, we found a strong left > right hemispheric asymmetry in aGMd that was, on average, less marked in the 40 left-handed subjects. Apparent GMd in the pCST increased with age in adolescent males but not females, and this was particularly related to rising plasma levels of testosterone in male adolescents. This finding is compatible with the idea that testosterone influences axonal calibre rather than myelination. The third prediction, namely that of a relationship between age-related changes in manual skills and maturation of the pCST, was not confirmed. We conclude that the leftward asymmetry of the pCST may reflect an early established asymmetry in the number of large corticomotoneuronal fibres in the pCST.
Collapse
Affiliation(s)
- Pierre-Yves Hervé
- Brain and Body Centre, University of Nottingham, Nottingham, United Kingdom.
| | | | | | | | | | | | | | | | | |
Collapse
|