1
|
Daniels N, Bindoff AD, Vickers JC, King AE, Collins JM. Vulnerability of neurofilament-expressing neurons in frontotemporal dementia. Mol Cell Neurosci 2024; 131:103974. [PMID: 39369804 DOI: 10.1016/j.mcn.2024.103974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 09/23/2024] [Accepted: 09/30/2024] [Indexed: 10/08/2024] Open
Abstract
Frontotemporal dementia (FTD) is an umbrella term for several early onset dementias, that are caused by frontotemporal lobar degeneration (FTLD), which involves the atrophy of the frontal and temporal lobes of the brain. Neuron loss in the frontal and temporal lobes is a characteristic feature of FTLD, however the selective vulnerability of different neuronal populations in this group of diseases is not fully understood. Neurofilament-expressing neurons have been shown to be selectively vulnerable in other neurodegenerative diseases, including Alzheimer's disease and amyotrophic lateral sclerosis, therefore we sought to investigate whether this neuronal population is vulnerable in FTLD. We also examined whether neuronal sub-type vulnerability differed between FTLD with TDP-43 inclusions (FTLD-TDP) and FTLD with tau inclusions (FTLD-Tau). Post-mortem human tissue from the superior frontal gyrus (SFG) of FTLD-TDP (n = 15), FTLD-Tau (n = 8) and aged Control cases (n = 6) was immunolabelled using antibodies against non-phosphorylated neurofilaments (SMI32 antibody), calretinin and NeuN, to explore neuronal cell loss. The presence of non-phosphorylated neurofilament immunolabelling in axons of the SFG white matter was also quantified as a measure of axon pathology, as axonal neurofilaments are normally phosphorylated. We demonstrate the selective loss of neurofilament-expressing neurons in both FTLD-TDP and FTLD-Tau cases compared to aged Controls. We also show that non-phosphorylated neurofilament axonal pathology in the SFG white matter was associated with increasing age, but not FTLD. This data suggests neurofilament-expressing neurons are vulnerable in both FTLD-TDP and FTLD-Tau.
Collapse
Affiliation(s)
- Nina Daniels
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, Tasmania, Australia.
| | - Aidan D Bindoff
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, Tasmania, Australia
| | - James C Vickers
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, Tasmania, Australia
| | - Anna E King
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, Tasmania, Australia
| | - Jessica M Collins
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, Tasmania, Australia
| |
Collapse
|
2
|
Calabrese M, Preziosa P, Scalfari A, Colato E, Marastoni D, Absinta M, Battaglini M, De Stefano N, Di Filippo M, Hametner S, Howell OW, Inglese M, Lassmann H, Martin R, Nicholas R, Reynolds R, Rocca MA, Tamanti A, Vercellino M, Villar LM, Filippi M, Magliozzi R. Determinants and Biomarkers of Progression Independent of Relapses in Multiple Sclerosis. Ann Neurol 2024; 96:1-20. [PMID: 38568026 DOI: 10.1002/ana.26913] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 01/04/2024] [Accepted: 02/15/2024] [Indexed: 06/20/2024]
Abstract
Clinical, pathological, and imaging evidence in multiple sclerosis (MS) suggests that a smoldering inflammatory activity is present from the earliest stages of the disease and underlies the progression of disability, which proceeds relentlessly and independently of clinical and radiological relapses (PIRA). The complex system of pathological events driving "chronic" worsening is likely linked with the early accumulation of compartmentalized inflammation within the central nervous system as well as insufficient repair phenomena and mitochondrial failure. These mechanisms are partially lesion-independent and differ from those causing clinical relapses and the formation of new focal demyelinating lesions; they lead to neuroaxonal dysfunction and death, myelin loss, glia alterations, and finally, a neuronal network dysfunction outweighing central nervous system (CNS) compensatory mechanisms. This review aims to provide an overview of the state of the art of neuropathological, immunological, and imaging knowledge about the mechanisms underlying the smoldering disease activity, focusing on possible early biomarkers and their translation into clinical practice. ANN NEUROL 2024;96:1-20.
Collapse
Affiliation(s)
- Massimiliano Calabrese
- Department of Neurosciences and Biomedicine and Movement, The Multiple Sclerosis Center of University Hospital of Verona, Verona, Italy
| | - Paolo Preziosa
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Antonio Scalfari
- Centre of Neuroscience, Department of Medicine, Imperial College, London, UK
| | - Elisa Colato
- Department of Neurosciences and Biomedicine and Movement, The Multiple Sclerosis Center of University Hospital of Verona, Verona, Italy
| | - Damiano Marastoni
- Department of Neurosciences and Biomedicine and Movement, The Multiple Sclerosis Center of University Hospital of Verona, Verona, Italy
| | - Martina Absinta
- Translational Neuropathology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Marco Battaglini
- Siena Imaging S.r.l., Siena, Italy
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Nicola De Stefano
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Massimiliano Di Filippo
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Simon Hametner
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Owain W Howell
- Institute of Life Sciences, Swansea University Medical School, Swansea, UK
| | - Matilde Inglese
- Dipartimento di neuroscienze, riabilitazione, oftalmologia, genetica e scienze materno-infantili - DINOGMI, University of Genova, Genoa, Italy
| | - Hans Lassmann
- Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Roland Martin
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
- Therapeutic Design Unit, Center for Molecular Medicine, Department of Clinical Neurosciences, Karolinska Institutet, Stockholm, Sweden
- Cellerys AG, Schlieren, Switzerland
| | - Richard Nicholas
- Department of Brain Sciences, Faculty of Medicine, Burlington Danes, Imperial College London, London, UK
| | - Richard Reynolds
- Division of Neuroscience, Department of Brain Sciences, Imperial College London, London, UK
| | - Maria A Rocca
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Agnese Tamanti
- Department of Neurosciences and Biomedicine and Movement, The Multiple Sclerosis Center of University Hospital of Verona, Verona, Italy
| | - Marco Vercellino
- Multiple Sclerosis Center & Neurologia I U, Department of Neuroscience, University Hospital AOU Città della Salute e della Scienza di Torino, Turin, Italy
| | - Luisa Maria Villar
- Department of Immunology, Ramon y Cajal University Hospital. IRYCIS. REI, Madrid, Spain
| | - Massimo Filippi
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
- Neurorehabilitation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurophysiology Service, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Roberta Magliozzi
- Department of Neurosciences and Biomedicine and Movement, The Multiple Sclerosis Center of University Hospital of Verona, Verona, Italy
| |
Collapse
|
3
|
Brotherton EJ, Sabapathy S, Heshmat S, Kavanagh JJ. Voluntary muscle activation in people with multiple sclerosis is reduced across a wide range of forces following maximal effort-fatiguing contractions. J Neurophysiol 2023; 130:1162-1173. [PMID: 37818597 DOI: 10.1152/jn.00146.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 10/04/2023] [Accepted: 10/04/2023] [Indexed: 10/12/2023] Open
Abstract
Although multiple sclerosis (MS) is frequently associated with motor impairment, little is known about how muscle activation is affected with MS. The aim of this study was to use transcranial magnetic stimulation (TMS) and motor nerve stimulation to investigate voluntary muscle activation in people with MS across a range of contraction forces. Ten people with MS (39 ± 7 yr) and 10 healthy controls (40 ± 5 yr) performed elbow flexions at target contraction forces of 25%, 50%, 75%, 90%, and 100% maximal voluntary contraction (MVC) while electromyography (EMG) of the biceps brachii was recorded. Sustained elbow flexion MVCs were then performed until force declined to 60% of baseline MVC, where the target contraction forces were again examined but after the sustained MVC. Following the sustained MVC, there was a reduction in biceps EMG amplitude (P < 0.01) and motor cortical voluntary activation (P < 0.01) for the MS group across all contraction intensities. There was also an increase in the rate of torque development for motor nerve-resting twitches in the MS group following the sustained MVC (P = 0.03). Despite the MS group reporting higher fatigue severity scale scores (P < 0.01), disease duration was a better predictor of muscle activation for the MS group (r = -0.757, P = 0.01). These findings indicate that voluntary muscle activation is compromised in people with MS following maximal effort contractions, which may be associated with disease duration rather than self-reports of fatigue.NEW & NOTEWORTHY We use transcranial magnetic stimulation to demonstrate that people with relapsing-remitting multiple sclerosis (MS) have a reduced ability to activate muscles following maximal effort-fatiguing contractions. A reduced ability to activate the elbow flexor muscles after a fatiguing contraction was associated with disease duration and not self-reported levels of fatigue.
Collapse
Affiliation(s)
- Emily J Brotherton
- Neural Control of Movement Laboratory, Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland, Australia
| | - Surendran Sabapathy
- Discipline of Exercise & Sport, School of Health Sciences and Social Work, Griffith University, Gold Coast, Queensland, Australia
| | - Saman Heshmat
- Department of Neurology, Gold Coast University Hospital, Gold Coast, Queensland, Australia
| | - Justin J Kavanagh
- Neural Control of Movement Laboratory, Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland, Australia
| |
Collapse
|
4
|
Toplu N, Oğuzoğlu TÇ. Caprine arthritis encephalitis virus-induced apoptosis associated with brain lesions in naturally infected kids. J Comp Pathol 2023; 206:36-43. [PMID: 37797470 DOI: 10.1016/j.jcpa.2023.08.008] [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: 03/07/2023] [Revised: 06/19/2023] [Accepted: 08/31/2023] [Indexed: 10/07/2023]
Abstract
Acute demyelinating leucoencephalomyelitis was the most conspicuous microscopic change in the brain and spinal cord of kids infected with caprine arthritis encephalitis virus (CAEV). TUNEL positivity and labelling of anti-bax and anti-caspases-3, -8 and -9 were found in a distinct population of glial cells, mainly at the edges of the demyelinated plaques and perivascular areas and, to a lesser extent, in neurons. Double labelling revealed that most of these apoptotic cells in the demyelinated plaques were astrocytes and a few were oligodendroglia. In contrast, expression of bcl-2, an anti-apoptotic protein, was found mainly in neurons of the brainstem and cerebellum and motor neurons of the spinal cord, but was restricted in glial cells. These results suggest that apoptosis plays an important role in the pathogenesis of CAE demyelinating encephalitis.
Collapse
Affiliation(s)
- Nihat Toplu
- Department of Pathology, Faculty of Veterinary Medicine, University of Aydın Adnan Menderes, 09016-Isikli, Aydin, Turkiye.
| | - Tuba Ç Oğuzoğlu
- Department of Virology, Faculty of Veterinary Medicine, University of Ankara, Diskapi, 06110 Ankara, Turkiye
| |
Collapse
|
5
|
Kapell H, Fazio L, Dyckow J, Schwarz S, Cruz-Herranz A, Mayer C, Campos J, D’Este E, Möbius W, Cordano C, Pröbstel AK, Gharagozloo M, Zulji A, Narayanan Naik V, Delank A, Cerina M, Müntefering T, Lerma-Martin C, Sonner JK, Sin JH, Disse P, Rychlik N, Sabeur K, Chavali M, Srivastava R, Heidenreich M, Fitzgerald KC, Seebohm G, Stadelmann C, Hemmer B, Platten M, Jentsch TJ, Engelhardt M, Budde T, Nave KA, Calabresi PA, Friese MA, Green AJ, Acuna C, Rowitch DH, Meuth SG, Schirmer L. Neuron-oligodendrocyte potassium shuttling at nodes of Ranvier protects against inflammatory demyelination. J Clin Invest 2023; 133:e164223. [PMID: 36719741 PMCID: PMC10065072 DOI: 10.1172/jci164223] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 01/27/2023] [Indexed: 02/01/2023] Open
Abstract
Multiple sclerosis (MS) is a progressive inflammatory demyelinating disease of the CNS. Increasing evidence suggests that vulnerable neurons in MS exhibit fatal metabolic exhaustion over time, a phenomenon hypothesized to be caused by chronic hyperexcitability. Axonal Kv7 (outward-rectifying) and oligodendroglial Kir4.1 (inward-rectifying) potassium channels have important roles in regulating neuronal excitability at and around the nodes of Ranvier. Here, we studied the spatial and functional relationship between neuronal Kv7 and oligodendroglial Kir4.1 channels and assessed the transcriptional and functional signatures of cortical and retinal projection neurons under physiological and inflammatory demyelinating conditions. We found that both channels became dysregulated in MS and experimental autoimmune encephalomyelitis (EAE), with Kir4.1 channels being chronically downregulated and Kv7 channel subunits being transiently upregulated during inflammatory demyelination. Further, we observed that pharmacological Kv7 channel opening with retigabine reduced neuronal hyperexcitability in human and EAE neurons, improved clinical EAE signs, and rescued neuronal pathology in oligodendrocyte-Kir4.1-deficient (OL-Kir4.1-deficient) mice. In summary, our findings indicate that neuron-OL compensatory interactions promoted resilience through Kv7 and Kir4.1 channels and identify pharmacological activation of nodal Kv7 channels as a neuroprotective strategy against inflammatory demyelination.
Collapse
Affiliation(s)
- Hannah Kapell
- Department of Neurology, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Luca Fazio
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster (UKM), Münster, Germany
- Department of Neurology, University of Düsseldorf, Dusseldorf, Germany
| | - Julia Dyckow
- Department of Neurology, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Sophia Schwarz
- Department of Neurology, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Andrés Cruz-Herranz
- Weill Institute for Neurosciences, Department of Neurology, UCSF, San Francisco, California, USA
| | - Christina Mayer
- Institute of Neuroimmunology and Multiple Sclerosis, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Joaquin Campos
- Chica and Heinz Schaller Research Group, Institute of Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Elisa D’Este
- Optical Microscopy Facility, Max Planck Institute for Medical Research, Heidelberg, Germany
| | - Wiebke Möbius
- Electron Microscopy Core Unit, Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
- Cluster of Excellence, “Multiscale Bioimaging: from Molecular Machines to Network of Excitable Cells” (MBExC), University of Göttingen, Göttingen, Germany
| | - Christian Cordano
- Weill Institute for Neurosciences, Department of Neurology, UCSF, San Francisco, California, USA
| | - Anne-Katrin Pröbstel
- Weill Institute for Neurosciences, Department of Neurology, UCSF, San Francisco, California, USA
- Neurologic Clinic and Policlinic and Research Center for Clinical Neuroimmunology and Neuroscience Basel, Departments of Medicine, Biomedicine, and Clinical Research, University Hospital of Basel, University of Basel, Basel, Switzerland
| | - Marjan Gharagozloo
- Departments of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Amel Zulji
- Department of Neurology, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Venu Narayanan Naik
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster (UKM), Münster, Germany
| | - Anna Delank
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster (UKM), Münster, Germany
| | - Manuela Cerina
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster (UKM), Münster, Germany
| | | | - Celia Lerma-Martin
- Department of Neurology, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Jana K. Sonner
- Chica and Heinz Schaller Research Group, Institute of Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Jung Hyung Sin
- Weill Institute for Neurosciences, Department of Neurology, UCSF, San Francisco, California, USA
| | - Paul Disse
- Institute for Genetics of Heart Diseases (IfGH), Cellular Electrophysiology and Molecular Biology, UKM, Münster, Germany
- University of Münster, Chembion, Münster, Germany
| | - Nicole Rychlik
- University of Münster, Chembion, Münster, Germany
- Institute of Physiology I, University of Münster, Münster, Germany
| | - Khalida Sabeur
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research and
- Department of Pediatrics, UCSF, San Francisco, California, USA
| | - Manideep Chavali
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research and
- Department of Pediatrics, UCSF, San Francisco, California, USA
| | - Rajneesh Srivastava
- Department of Neurology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Matthias Heidenreich
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP) and Max-Delbrück-Centrum für Molekulare Medizin (MDC), Berlin, Germany
| | - Kathryn C. Fitzgerald
- Departments of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Guiscard Seebohm
- Institute for Genetics of Heart Diseases (IfGH), Cellular Electrophysiology and Molecular Biology, UKM, Münster, Germany
| | - Christine Stadelmann
- Electron Microscopy Core Unit, Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
- Institute of Neuropathology, University Medical Center, Göttingen, Germany
| | - Bernhard Hemmer
- Department of Neurology, School of Medicine, Technical University of Munich, Munich, Germany
- Munich Cluster for Systems Neurology, Munich, Germany
| | - Michael Platten
- Department of Neurology, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
- DKTK Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), INF 280, Heidelberg, Germany
- Interdisciplinary Center for Neurosciences (IZN) and
- Mannheim Center for Translational Neuroscience and Institute for Innate Immunoscience, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Thomas J. Jentsch
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP) and Max-Delbrück-Centrum für Molekulare Medizin (MDC), Berlin, Germany
- Neurocure Cluster of Excellence, Charité University Medicine Berlin, Berlin, Germany
| | - Maren Engelhardt
- Mannheim Center for Translational Neuroscience and Institute for Innate Immunoscience, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
- Institute of Neuroanatomy, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
- Institute of Anatomy and Cell Biology, Johannes Kepler University Linz, Linz, Austria
| | - Thomas Budde
- Institute of Physiology I, University of Münster, Münster, Germany
| | - Klaus-Armin Nave
- Electron Microscopy Core Unit, Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Peter A. Calabresi
- Departments of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Manuel A. Friese
- Institute of Neuroimmunology and Multiple Sclerosis, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Ari J. Green
- Weill Institute for Neurosciences, Department of Neurology, UCSF, San Francisco, California, USA
- Department of Ophthalmology, UCSF, San Francisco, California, USA
| | - Claudio Acuna
- Chica and Heinz Schaller Research Group, Institute of Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - David H. Rowitch
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research and
- Department of Pediatrics, UCSF, San Francisco, California, USA
- Wellcome Trust–Medical Research Council Stem Cell Institute and
- Department of Paediatrics, University of Cambridge, Cambridge, United Kingdom
- Department of Neurosurgery, UCSF, San Francisco, California, USA
| | - Sven G. Meuth
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster (UKM), Münster, Germany
- Department of Neurology, University of Düsseldorf, Dusseldorf, Germany
| | - Lucas Schirmer
- Department of Neurology, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
- Interdisciplinary Center for Neurosciences (IZN) and
- Mannheim Center for Translational Neuroscience and Institute for Innate Immunoscience, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
6
|
Bian Z, Hu X, Liu X, Yu H, Bian Y, Sun H, Fukui Y, Morihara R, Ishiura H, Yamashita T. Protective Effects of Rivaroxaban on White Matter Integrity and Remyelination in a Mouse Model of Alzheimer's Disease Combined with Cerebral Hypoperfusion. J Alzheimers Dis 2023; 96:609-622. [PMID: 37840489 DOI: 10.3233/jad-230413] [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] [Indexed: 10/17/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is characterized by cognitive dysfunction and memory loss that is accompanied by pathological changes to white matter. Some clinical and animal research revealed that AD combined with chronic cerebral hypoperfusion (CCH) exacerbates AD progression by inducing blood-brain barrier dysfunction and fibrinogen deposition. Rivaroxaban, an anticoagulant, has been shown to reduce the rates of dementia in atrial fibrillation patients, but its effects on white matter and the underlying mechanisms are unclear. OBJECTIVE The main purpose of this study was to explore the therapeutic effect of rivaroxaban on the white matter of AD+CCH mice. METHODS In this study, the therapeutic effects of rivaroxaban on white matter in a mouse AD+CCH model were investigated to explore the potential mechanisms involving fibrinogen deposition, inflammation, and oxidative stress on remyelination in white matter. RESULTS The results indicate that rivaroxaban significantly attenuated fibrinogen deposition, fibrinogen-related microglia activation, oxidative stress, and enhanced demyelination in AD+CCH mice, leading to improved white matter integrity, reduced axonal damage, and restored myelin loss. CONCLUSIONS These findings suggest that long-term administration of rivaroxaban might reduce the risk of dementia.
Collapse
Affiliation(s)
- Zhihong Bian
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Xinran Hu
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Xia Liu
- Department of Neurology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haibo Yu
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Yuting Bian
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Hongming Sun
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Yusuke Fukui
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Ryuta Morihara
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Hiroyuki Ishiura
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Toru Yamashita
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| |
Collapse
|
7
|
Orekhova K, Selmanovic E, De Gasperi R, Gama Sosa MA, Wicinski B, Maloney B, Seifert A, Alipour A, Balchandani P, Gerussi T, Graïc JM, Centelleghe C, Di Guardo G, Mazzariol S, Hof PR. Multimodal Assessment of Bottlenose Dolphin Auditory Nuclei Using 7-Tesla MRI, Immunohistochemistry and Stereology. Vet Sci 2022; 9:vetsci9120692. [PMID: 36548853 PMCID: PMC9781543 DOI: 10.3390/vetsci9120692] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/05/2022] [Accepted: 12/09/2022] [Indexed: 12/14/2022] Open
Abstract
The importance of assessing neurochemical processes in the cetacean brain as a tool for monitoring their cognitive health and to indirectly model human neurodegenerative conditions is increasingly evident, although available data are largely semiquantitative. High-resolution MRI for post-mortem brains and stereology allow for quantitative assessments of the cetacean brain. In this study, we scanned two brains of bottlenose dolphins in a 7-Tesla (7T) MR scanner and assessed the connectivity of the inferior colliculi and ventral cochlear nuclei using diffusion tensor imaging (DTI). Serial thick sections were investigated stereologically in one of the dolphins to generate rigorous quantitative estimates of identifiable cell types according to their morphology and expression of molecular markers, yielding reliable cell counts with most coefficients of error <10%. Fibronectin immunoreactivity in the dolphin resembled the pattern in a human chronic traumatic encephalopathy brain, suggesting that neurochemical compensation for insults such as hypoxia may constitute a noxious response in humans, while being physiological in dolphins. These data contribute to a growing body of knowledge on the morphological and neurochemical properties of the dolphin brain and highlight a stereological and neuroimaging workflow that may enable quantitative and translational assessment of pathological processes in the dolphin brain in the future.
Collapse
Affiliation(s)
- Ksenia Orekhova
- Department of Comparative Biomedicine and Food Science, University of Padova AGRIPOLIS, Viale dell’Università 16, 35020 Legnaro, Italy
- Correspondence:
| | - Enna Selmanovic
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Rita De Gasperi
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, Bronx, New York, NY 10468, USA
| | - Miguel A. Gama Sosa
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- General Medical Research Service, James J. Peters Department of Veterans Affairs Medical Center, Bronx, New York, NY 10468, USA
| | - Bridget Wicinski
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Brigid Maloney
- Laboratory of Neurogenetics of Vocal Learning, Rockefeller University, New York, NY 10065, USA
| | - Alan Seifert
- Department of Radiology, BioMedical Engineering and Imaging Institute (BMEII), Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Akbar Alipour
- Department of Radiology, BioMedical Engineering and Imaging Institute (BMEII), Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Priti Balchandani
- Department of Radiology, BioMedical Engineering and Imaging Institute (BMEII), Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Tommaso Gerussi
- Department of Comparative Biomedicine and Food Science, University of Padova AGRIPOLIS, Viale dell’Università 16, 35020 Legnaro, Italy
| | - Jean-Marie Graïc
- Department of Comparative Biomedicine and Food Science, University of Padova AGRIPOLIS, Viale dell’Università 16, 35020 Legnaro, Italy
| | - Cinzia Centelleghe
- Department of Comparative Biomedicine and Food Science, University of Padova AGRIPOLIS, Viale dell’Università 16, 35020 Legnaro, Italy
| | - Giovanni Di Guardo
- Faculty of Veterinary Medicine, University of Teramo, 64100 Teramo, Italy
| | - Sandro Mazzariol
- Department of Comparative Biomedicine and Food Science, University of Padova AGRIPOLIS, Viale dell’Università 16, 35020 Legnaro, Italy
| | - Patrick R. Hof
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
8
|
Kor DZL, Jbabdi S, Huszar IN, Mollink J, Tendler BC, Foxley S, Wang C, Scott C, Smart A, Ansorge O, Pallebage-Gamarallage M, Miller KL, Howard AFD. An automated pipeline for extracting histological stain area fraction for voxelwise quantitative MRI-histology comparisons. Neuroimage 2022; 264:119726. [PMID: 36368503 PMCID: PMC10933753 DOI: 10.1016/j.neuroimage.2022.119726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 10/27/2022] [Accepted: 10/31/2022] [Indexed: 11/11/2022] Open
Abstract
The acquisition of MRI and histology in the same post-mortem tissue sample enables direct correlation between MRI and histologically-derived parameters. However, there still lacks a standardised automated pipeline to process histology data, with most studies relying on manual intervention. Here, we introduce an automated pipeline to extract a quantitative histological measure for staining density (stain area fraction, SAF) from multiple immunohistochemical (IHC) stains. The pipeline is designed to directly address key IHC artefacts related to tissue staining and slide digitisation. Here, the pipeline was applied to post-mortem human brain data from multiple subjects, relating MRI parameters (FA, MD, RD, AD, R2*, R1) to IHC slides stained for myelin, neurofilaments, microglia and activated microglia. Utilising high-quality MRI-histology co-registrations, we then performed whole-slide voxelwise comparisons (simple correlations, partial correlations and multiple regression analyses) between multimodal MRI- and IHC-derived parameters. The pipeline was found to be reproducible, robust to artefacts and generalisable across multiple IHC stains. Our partial correlation results suggest that some simple MRI-SAF correlations should be interpreted with caution, due to the co-localisation of other tissue features (e.g., myelin and neurofilaments). Further, we find activated microglia-a generic biomarker of inflammation-to consistently be the strongest predictor of high DTI FA and low RD, which may suggest sensitivity of diffusion MRI to aspects of neuroinflammation related to microglial activation, even after accounting for other microstructural changes (demyelination, axonal loss and general microglia infiltration). Together, these results show the utility of this approach in carefully curating IHC data and performing multimodal analyses to better understand microstructural relationships with MRI.
Collapse
Affiliation(s)
- Daniel Z L Kor
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences, University of Oxford, Headington, Oxford OX3 9DU, , United Kingdom.
| | - Saad Jbabdi
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences, University of Oxford, Headington, Oxford OX3 9DU, , United Kingdom
| | - Istvan N Huszar
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences, University of Oxford, Headington, Oxford OX3 9DU, , United Kingdom
| | - Jeroen Mollink
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences, University of Oxford, Headington, Oxford OX3 9DU, , United Kingdom
| | - Benjamin C Tendler
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences, University of Oxford, Headington, Oxford OX3 9DU, , United Kingdom
| | - Sean Foxley
- Department of Radiology, University of Chicago, Chicago, IL, United States of America
| | - Chaoyue Wang
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences, University of Oxford, Headington, Oxford OX3 9DU, , United Kingdom
| | - Connor Scott
- Academic Unit of Neuropathology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Adele Smart
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences, University of Oxford, Headington, Oxford OX3 9DU, , United Kingdom; Academic Unit of Neuropathology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Olaf Ansorge
- Academic Unit of Neuropathology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Menuka Pallebage-Gamarallage
- Academic Unit of Neuropathology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Karla L Miller
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences, University of Oxford, Headington, Oxford OX3 9DU, , United Kingdom
| | - Amy F D Howard
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences, University of Oxford, Headington, Oxford OX3 9DU, , United Kingdom
| |
Collapse
|
9
|
Purkinje cell vulnerability induced by diffuse traumatic brain injury is linked to disruption of long-range neuronal circuits. Acta Neuropathol Commun 2022; 10:129. [PMID: 36064443 PMCID: PMC9446851 DOI: 10.1186/s40478-022-01435-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 08/22/2022] [Indexed: 11/10/2022] Open
Abstract
Cerebellar dysfunction is commonly observed following traumatic brain injury (TBI). While direct impact to the cerebellum by TBI is rare, cerebellar pathology may be caused by indirect injury via cortico-cerebellar pathways. To address the hypothesis that degeneration of Purkinje cells (PCs), which constitute the sole output from the cerebellum, is linked to long-range axonal injury and demyelination, we used the central fluid percussion injury (cFPI) model of widespread traumatic axonal injury in mice. Compared to controls, TBI resulted in early PC loss accompanied by alterations in the size of pinceau synapses and levels of non-phosphorylated neurofilament in PCs. A combination of vDISCO tissue clearing technique and immunohistochemistry for vesicular glutamate transporter type 2 show that diffuse TBI decreased mossy and climbing fiber synapses on PCs. At 2 days post-injury, numerous axonal varicosities were found in the cerebellum supported by fractional anisotropy measurements using 9.4 T MRI. The disruption and demyelination of the cortico-cerebellar circuits was associated with poor performance of brain-injured mice in the beam-walk test. Despite a lack of direct input from the injury site to the cerebellum, these findings argue for novel long-range mechanisms causing Purkinje cell injury that likely contribute to cerebellar dysfunction after TBI.
Collapse
|
10
|
Gagnon J, Pi L, Ryals M, Wan Q, Hu W, Ouyang Z, Zhang B, Li K. Recommendations of scRNA-seq Differential Gene Expression Analysis Based on Comprehensive Benchmarking. LIFE (BASEL, SWITZERLAND) 2022; 12:life12060850. [PMID: 35743881 PMCID: PMC9225332 DOI: 10.3390/life12060850] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 05/31/2022] [Accepted: 06/04/2022] [Indexed: 12/13/2022]
Abstract
To guide analysts to select the right tool and parameters in differential gene expression analyses of single-cell RNA sequencing (scRNA-seq) data, we developed a novel simulator that recapitulates the data characteristics of real scRNA-seq datasets while accounting for all the relevant sources of variation in a multi-subject, multi-condition scRNA-seq experiment: the cell-to-cell variation within a subject, the variation across subjects, the variability across cell types, the mean/variance relationship of gene expression across genes, library size effects, group effects, and covariate effects. By applying it to benchmark 12 differential gene expression analysis methods (including cell-level and pseudo-bulk methods) on simulated multi-condition, multi-subject data of the 10x Genomics platform, we demonstrated that methods originating from the negative binomial mixed model such as glmmTMB and NEBULA-HL outperformed other methods. Utilizing NEBULA-HL in a statistical analysis pipeline for single-cell analysis will enable scientists to better understand the cell-type-specific transcriptomic response to disease or treatment effects and to discover new drug targets. Further, application to two real datasets showed the outperformance of our differential expression (DE) pipeline, with unified findings of differentially expressed genes (DEG) and a pseudo-time trajectory transcriptomic result. In the end, we made recommendations for filtering strategies of cells and genes based on simulation results to achieve optimal experimental goals.
Collapse
Affiliation(s)
- Jake Gagnon
- Analytics and Data Sciences, Biogen, Inc., 225 Binney St., Cambridge, MA 02142, USA;
| | - Lira Pi
- PharmaLex, 1700 District Ave., Burlington, MA 01803, USA; (L.P.); (M.R.); (Q.W.)
| | - Matthew Ryals
- PharmaLex, 1700 District Ave., Burlington, MA 01803, USA; (L.P.); (M.R.); (Q.W.)
| | - Qingwen Wan
- PharmaLex, 1700 District Ave., Burlington, MA 01803, USA; (L.P.); (M.R.); (Q.W.)
| | - Wenxing Hu
- Research Department, Biogen, Inc., 225 Binney St., Cambridge, MA 02142, USA;
| | - Zhengyu Ouyang
- BioInfoRx, Inc., 510 Charmany Dr., Suite 275A, Madison, WI 53719, USA;
| | - Baohong Zhang
- Research Department, Biogen, Inc., 225 Binney St., Cambridge, MA 02142, USA;
- Correspondence: (B.Z.); (K.L.)
| | - Kejie Li
- Research Department, Biogen, Inc., 225 Binney St., Cambridge, MA 02142, USA;
- Correspondence: (B.Z.); (K.L.)
| |
Collapse
|
11
|
Linnerbauer M, Lößlein L, Vandrey O, Tsaktanis T, Beer A, Naumann UJ, Panier F, Beyer T, Nirschl L, Kuramatsu JB, Winkler J, Quintana FJ, Rothhammer V. Intranasal delivery of a small-molecule ErbB inhibitor promotes recovery from acute and late-stage CNS inflammation. JCI Insight 2022; 7:154824. [PMID: 35393953 PMCID: PMC9057609 DOI: 10.1172/jci.insight.154824] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 02/18/2022] [Indexed: 11/17/2022] Open
Abstract
Multiple sclerosis (MS) is an autoimmune inflammatory disease of the CNS that is characterized by demyelination and axonal degeneration. Although several established treatments reduce relapse burden, effective treatments to halt chronic progression are scarce. Single-cell transcriptomic studies in MS and its animal models have described astrocytes and their spatial and functional heterogeneity as important cellular determinants of chronic disease. We combined CNS single-cell transcriptome data and small-molecule screens in primary mouse and human astrocytes to identify glial interactions, which could be targeted by repurposing FDA-approved small-molecule modulators for the treatment of acute and late-stage CNS inflammation. Using hierarchical in vitro and in vivo validation studies, we demonstrate that among selected pathways, blockade of ErbB by the tyrosine kinase inhibitor afatinib efficiently mitigates proinflammatory astrocyte polarization and promotes tissue-regenerative functions. We found that i.n. delivery of afatinib during acute and late-stage CNS inflammation ameliorates disease severity by reducing monocyte infiltration and axonal degeneration while increasing oligodendrocyte proliferation. We used unbiased screening approaches of astrocyte interactions to identify ErbB signaling and its modulation by afatinib as a potential therapeutic strategy for acute and chronic stages of autoimmune CNS inflammation.
Collapse
Affiliation(s)
- Mathias Linnerbauer
- Department of Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Lena Lößlein
- Department of Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Oliver Vandrey
- Department of Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Thanos Tsaktanis
- Department of Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Alexander Beer
- Department of Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Ulrike J Naumann
- Department of Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Franziska Panier
- Department of Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Tobias Beyer
- Department of Neurology, Rechts der Isar Hospital, Technical University of Munich, Munich, Germany
| | - Lucy Nirschl
- Department of Neurology, Rechts der Isar Hospital, Technical University of Munich, Munich, Germany
| | - Joji B Kuramatsu
- Department of Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Jürgen Winkler
- Department of Molecular Neurology, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Francisco J Quintana
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Veit Rothhammer
- Department of Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
12
|
Alpaugh M, Masnata M, de Rus Jacquet A, Lepinay E, Denis HL, Saint-Pierre M, Davies P, Planel E, Cicchetti F. Passive immunization against phosphorylated tau improves features of Huntington's disease pathology. Mol Ther 2022; 30:1500-1522. [PMID: 35051614 PMCID: PMC9077324 DOI: 10.1016/j.ymthe.2022.01.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 11/05/2021] [Accepted: 01/12/2022] [Indexed: 01/07/2023] Open
Abstract
Huntington's disease is classically described as a neurodegenerative disorder of monogenic aetiology. The disease is characterized by an abnormal polyglutamine expansion in the huntingtin gene, which drives the toxicity of the mutated form of the protein. However, accumulation of the microtubule-associated protein tau, which is involved in a number of neurological disorders, has also been observed in patients with Huntington's disease. In order to unravel the contribution of tau hyperphosphorylation to hallmark features of Huntington's disease, we administered weekly intraperitoneal injections of the anti-tau pS202 CP13 monoclonal antibody to zQ175 mice and characterized the resulting behavioral and biochemical changes. After 12 weeks of treatment, motor impairments, cognitive performance and general health were improved in zQ175 mice along with a significant reduction in hippocampal pS202 tau levels. Despite the lack of effect of CP13 on neuronal markers associated with Huntington's disease pathology, tau-targeting enzymes and gliosis, CP13 was shown to directly impact mutant huntingtin aggregation such that brain levels of amyloid fibrils and huntingtin oligomers were decreased, while larger huntingtin protein aggregates were increased. Investigation of CP13 treatment of Huntington's disease patient-derived induced pluripotent stem cells (iPSCs) revealed a reduction in pS202 levels in differentiated cortical neurons and a rescue of neurite length. Collectively, these findings suggest that attenuating tau pathology could mitigate behavioral and molecular hallmarks associated with Huntington's disease.
Collapse
Affiliation(s)
- Melanie Alpaugh
- Centre de Recherche du CHU de Québec - Université Laval, Axe Neurosciences, Québec, QC G1V 4G2, Canada; Département de Psychiatrie & Neurosciences, Université Laval, Québec, QC G1K 0A6, Canada
| | - Maria Masnata
- Centre de Recherche du CHU de Québec - Université Laval, Axe Neurosciences, Québec, QC G1V 4G2, Canada; Département de Psychiatrie & Neurosciences, Université Laval, Québec, QC G1K 0A6, Canada
| | - Aurelie de Rus Jacquet
- Centre de Recherche du CHU de Québec - Université Laval, Axe Neurosciences, Québec, QC G1V 4G2, Canada; Département de Psychiatrie & Neurosciences, Université Laval, Québec, QC G1K 0A6, Canada
| | - Eva Lepinay
- Centre de Recherche du CHU de Québec - Université Laval, Axe Neurosciences, Québec, QC G1V 4G2, Canada; Département de Psychiatrie & Neurosciences, Université Laval, Québec, QC G1K 0A6, Canada
| | - Hélèna L Denis
- Centre de Recherche du CHU de Québec - Université Laval, Axe Neurosciences, Québec, QC G1V 4G2, Canada; Département de Psychiatrie & Neurosciences, Université Laval, Québec, QC G1K 0A6, Canada
| | - Martine Saint-Pierre
- Département de Psychiatrie & Neurosciences, Université Laval, Québec, QC G1K 0A6, Canada
| | - Peter Davies
- Albert Einstein College of Medicine, Bronx, NY, USA
| | - Emmanuel Planel
- Centre de Recherche du CHU de Québec - Université Laval, Axe Neurosciences, Québec, QC G1V 4G2, Canada; Département de Psychiatrie & Neurosciences, Université Laval, Québec, QC G1K 0A6, Canada
| | - Francesca Cicchetti
- Centre de Recherche du CHU de Québec - Université Laval, Axe Neurosciences, Québec, QC G1V 4G2, Canada; Département de Psychiatrie & Neurosciences, Université Laval, Québec, QC G1K 0A6, Canada.
| |
Collapse
|
13
|
Tipton PW, Kenney-Jung D, Rush BK, Middlebrooks EH, Nascene D, Singh B, Holtan S, Ayala E, Broderick DF, Lund T, Wszolek ZK. Treatment of CSF1R-Related Leukoencephalopathy: Breaking New Ground. Mov Disord 2021; 36:2901-2909. [PMID: 34329526 DOI: 10.1002/mds.28734] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 06/02/2021] [Accepted: 07/13/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Colony-stimulating factor-1 receptor (CSF1R)-related leukoencephalopathy is a rapidly progressive neurodegenerative disease for which there is currently no cure. Hematopoietic stem cell transplantation (HSCT) has been proposed as a disease-modifying treatment. OBJECTIVE The objective of this study was to determine the effect of HSCT on disease progression. METHODS We collected all available clinical data from a cohort of 7 patients with CSF1R-related leukoencephalopathy who underwent HSCT at our institutions. Clinical data included detailed neurological examination by a board-certified neurologist, serial cognitive screens, formal neuropsychological evaluations, and serial brain magnetic resonance imaging (MRI). RESULTS Our patients had an average disease duration of 27.6 months at the time of transplant, and we have 87 months of total posttransplant follow-up time (median, 11; range, 2-27). One patient died in the periprocedural period. The remaining patients showed a variable response to treatment, with 6 of 7 patients trending toward stabilization on motor examination, cognitive scores, and/or MRI abnormalities, especially with white matter lesion burden. CONCLUSIONS This is the largest series of patients with CSF1R-related leukoencephalopathy receiving HSCT. We conclude that HSCT can stabilize the disease in some patients. Variability in patient responsiveness suggests that measures of disease heterogeneity and severity need to be considered when evaluating a patient's candidacy for transplant. HSCT appears to be the first disease-modifying therapy for CSF1R-related leukoencephalopathy. This milestone may serve as a foothold toward better understanding the disease's pathomechanism, thus providing new opportunities for better disease-specific therapies. © 2021 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Philip W Tipton
- Department of Neurology, Mayo Clinic, Jacksonville, Florida, USA
| | - Daniel Kenney-Jung
- Department of Neurology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Beth K Rush
- Department of Psychiatry & Psychology, Mayo Clinic, Jacksonville, Florida, USA
| | | | - David Nascene
- Department of Radiology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Balvindar Singh
- Department of Neurology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Shernan Holtan
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Ernesto Ayala
- Department of Hematology Oncology, Mayo Clinic, Jacksonville, Florida, USA
| | | | - Troy Lund
- Department of Pediatrics, Division of Blood and Marrow Transplant, University of Minnesota, Minneapolis, Minnesota, USA
| | | |
Collapse
|
14
|
Kang Y, Pandya S, Zinger N, Michaelson N, Gauthier SA. Longitudinal change in TSPO PET imaging in progressive multiple sclerosis. Ann Clin Transl Neurol 2021; 8:1755-1759. [PMID: 34310086 PMCID: PMC8351399 DOI: 10.1002/acn3.51431] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 05/31/2021] [Accepted: 07/06/2021] [Indexed: 11/06/2022] Open
Abstract
The objective of this pilot study was to assess a 2-year change in innate immune burden in 15 progressive multiple sclerosis (MS) patients using PK11195-PET. Sixteen age-matched healthy controls (HC) were included for baseline comparison. PK11195 uptake was higher in MS patients compared to HC within normal-appearing white matter (NAWM) and multiple gray matter regions. In patients, PK11195 uptake increased in NAWM (p = 0.01), cortex (p = 0.04), thalamus (p = 0.04), and putamen (p = 0.02) at 12 months. Among patients remaining at 24 months, there was no further increase in PK11195. Our data suggest that innate immune activity may increase over time in patients with progressive MS.
Collapse
Affiliation(s)
- Yeona Kang
- Department of Mathematics, Howard University, Washington, District of Columbia, USA.,Department of Radiology, Weill Cornell Medicine, New York, New York, USA
| | - Sneha Pandya
- Department of Radiology, Weill Cornell Medicine, New York, New York, USA
| | - Nicole Zinger
- Department of Neurology, Weill Cornell Medicine, New York, New York, USA
| | - Nara Michaelson
- Department of Neurology, Weill Cornell Medicine, New York, New York, USA
| | - Susan A Gauthier
- Department of Radiology, Weill Cornell Medicine, New York, New York, USA.,Department of Neurology, Weill Cornell Medicine, New York, New York, USA.,Feil Brain and Mind Institute, Weill Cornell Medicine, New York, New York, USA
| |
Collapse
|
15
|
Bhargava P, Kim S, Reyes AA, Grenningloh R, Boschert U, Absinta M, Pardo C, Van Zijl P, Zhang J, Calabresi PA. Imaging meningeal inflammation in CNS autoimmunity identifies a therapeutic role for BTK inhibition. Brain 2021; 144:1396-1408. [PMID: 33724342 DOI: 10.1093/brain/awab045] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 11/17/2020] [Accepted: 12/03/2020] [Indexed: 02/06/2023] Open
Abstract
Leptomeningeal inflammation in multiple sclerosis is associated with worse clinical outcomes and greater cortical pathology. Despite progress in identifying this process in multiple sclerosis patients using post-contrast fluid-attenuated inversion recovery imaging, early trials attempting to target meningeal inflammation have been unsuccessful. There is a lack of appropriate model systems to screen potential therapeutic agents targeting meningeal inflammation. We utilized ultra-high field (11.7 T) MRI to perform post-contrast imaging in SJL/J mice with experimental autoimmune encephalomyelitis induced via immunization with proteolipid protein peptide (PLP139-151) and complete Freund's adjuvant. Imaging was performed in both a cross-sectional and longitudinal fashion at time points ranging from 2 to 14 weeks post-immunization. Following imaging, we euthanized animals and collected tissue for pathological evaluation, which revealed dense cellular infiltrates corresponding to areas of contrast enhancement involving the leptomeninges. These areas of meningeal inflammation contained B cells (B220+), T cells (CD3+) and myeloid cells (Mac2+). We also noted features consistent with tertiary lymphoid tissue within these areas, namely the presence of peripheral node addressin-positive structures, C-X-C motif chemokine ligand-13 (CXCL13)-producing cells and FDC-M1+ follicular dendritic cells. In the cortex adjacent to areas of meningeal inflammation we identified astrocytosis, microgliosis, demyelination and evidence of axonal stress/damage. Since areas of meningeal contrast enhancement persisted over several weeks in longitudinal experiments, we utilized this model to test the effects of a therapeutic intervention on established meningeal inflammation. We randomized mice with evidence of meningeal contrast enhancement on MRI scans performed at 6 weeks post-immunization, to treatment with either vehicle or evobrutinib [a Bruton tyrosine kinase (BTK) inhibitor] for a period of 4 weeks. These mice underwent serial imaging; we examined the effect of treatment on the areas of meningeal contrast enhancement and noted a significant reduction in the evobrutinib group compared to vehicle (30% reduction versus 5% increase; P = 0.003). We used ultra-high field MRI to identify areas of meningeal inflammation and to track them over time in SJL/J mice with experimental autoimmune encephalomyelitis, and then used this model to identify BTK inhibition as a novel therapeutic approach to target meningeal inflammation. The results of this study provide support for future studies in multiple sclerosis patients with imaging evidence of meningeal inflammation.
Collapse
Affiliation(s)
- Pavan Bhargava
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sol Kim
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Arthur A Reyes
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | | | - Martina Absinta
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Carlos Pardo
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Peter Van Zijl
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Jiangyang Zhang
- Department of Radiology, New York University, New York, NY, USA
| | - Peter A Calabresi
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
16
|
Hares K, Kemp K, Loveless S, Rice CM, Scolding N, Tallantyre E, Robertson N, Wilkins A. KIF5A and the contribution of susceptibility genotypes as a predictive biomarker for multiple sclerosis. J Neurol 2021; 268:2175-2184. [PMID: 33484325 PMCID: PMC8179895 DOI: 10.1007/s00415-020-10373-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 12/11/2020] [Accepted: 12/14/2020] [Indexed: 12/01/2022]
Abstract
There is increasing interest in the development of multiple sclerosis (MS) biomarkers that reflect central nervous system tissue injury to determine prognosis. We aimed to assess the prognostic value of kinesin superfamily motor protein KIF5A in MS by measuring levels of KIF5A in cerebrospinal fluid (CSF) combined with analysis of single nucleotide polymorphisms (SNPs; rs12368653 and rs703842) located within a MS susceptibility gene locus at chromosome 12q13-14 region. Enzyme-linked immunosorbent assay was used to measure KIF5A in CSF obtained from two independent biobanks comprising non-inflammatory neurological disease controls (NINDC), clinically isolated syndrome (CIS) and MS cases. CSF KIF5A expression was significantly elevated in progressive MS cases compared with NINDCs, CIS and relapsing-remitting MS (RRMS). In addition, levels of KIF5A positively correlated with change in MS disease severity scores (EDSS, MSSS and ARMSSS), in RRMS patients who had documented disease progression at 2-year clinical follow-up. Copies of adenine risk alleles (AG/AA; rs12368653 and rs703842) corresponded with a higher proportion of individuals in relapse at the time of lumbar puncture (LP), higher use of disease-modifying therapies post LP and shorter MS duration. Our study suggests that CSF KIF5A has potential as a predictive biomarker in MS and further studies into the potential prognostic value of analysing MS susceptibility SNPs should be considered.
Collapse
Affiliation(s)
- Kelly Hares
- MS and Stem Cell Group, Institute of Clinical Neurosciences, Bristol Medical School: Translational Health Sciences, University of Bristol, Clinical Neurosciences Office, 1st Floor, Learning and Research Building, Southmead Hospital, Bristol, BS10 5NB, UK.
| | - K Kemp
- MS and Stem Cell Group, Institute of Clinical Neurosciences, Bristol Medical School: Translational Health Sciences, University of Bristol, Clinical Neurosciences Office, 1st Floor, Learning and Research Building, Southmead Hospital, Bristol, BS10 5NB, UK
| | - S Loveless
- Division of Psychological Medicine and Clinical Neuroscience, School of Medicine, Cardiff University, Cardiff, UK
| | - C M Rice
- MS and Stem Cell Group, Institute of Clinical Neurosciences, Bristol Medical School: Translational Health Sciences, University of Bristol, Clinical Neurosciences Office, 1st Floor, Learning and Research Building, Southmead Hospital, Bristol, BS10 5NB, UK
| | - N Scolding
- MS and Stem Cell Group, Institute of Clinical Neurosciences, Bristol Medical School: Translational Health Sciences, University of Bristol, Clinical Neurosciences Office, 1st Floor, Learning and Research Building, Southmead Hospital, Bristol, BS10 5NB, UK
| | - E Tallantyre
- Division of Psychological Medicine and Clinical Neuroscience, School of Medicine, Cardiff University, Cardiff, UK
| | - N Robertson
- Division of Psychological Medicine and Clinical Neuroscience, School of Medicine, Cardiff University, Cardiff, UK
| | - A Wilkins
- MS and Stem Cell Group, Institute of Clinical Neurosciences, Bristol Medical School: Translational Health Sciences, University of Bristol, Clinical Neurosciences Office, 1st Floor, Learning and Research Building, Southmead Hospital, Bristol, BS10 5NB, UK
| |
Collapse
|
17
|
Kleiter I, Ayzenberg I, Havla J, Lukas C, Penner IK, Stadelmann C, Linker RA. The transitional phase of multiple sclerosis: Characterization and conceptual framework. Mult Scler Relat Disord 2020; 44:102242. [DOI: 10.1016/j.msard.2020.102242] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 05/17/2020] [Accepted: 05/24/2020] [Indexed: 10/24/2022]
|
18
|
Teuber-Hanselmann S, Meinl E, Junker A. MicroRNAs in gray and white matter multiple sclerosis lesions: impact on pathophysiology. J Pathol 2020; 250:496-509. [PMID: 32073139 DOI: 10.1002/path.5399] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 02/05/2020] [Accepted: 02/11/2020] [Indexed: 12/12/2022]
Abstract
Multiple sclerosis (MS) is a chronic disease of the CNS, hallmarked by inflammation and demyelination. Early stages of the disease frequently show active lesions containing numerous foamy macrophages and inflammatory cells. Disease progression is highlighted by increasing numbers of mixed active/inactive or inactive lesions showing sparse inflammation and pronounced astrogliosis. Furthermore, gray matter lesions increase in number and extent during disease progression. MicroRNAs (miRNAs) comprise a group of several thousand (in humans more than 2000), small non-coding RNA molecules with a fundamental influence on about one-third of all protein-coding genes. Furthermore, miRNAs have been detected in body fluids, including spinal fluid, and they are assumed to participate in intercellular communications. Several studies have determined miRNA profiles from dissected white and gray matter lesions of autoptic MS patients. In this review, we summarize in detail the current knowledge of individual miRNAs in gray and white matter lesions of MS patients and present the concepts of MS tissue lesion development based on the altered miRNA profiles. © 2020 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
| | - Edgar Meinl
- Institute of Clinical Neuroimmunology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Andreas Junker
- Institute of Neuropathology, University Hospital Essen, Essen, Germany
| |
Collapse
|
19
|
Anaby D, Morozov D, Seroussi I, Hametner S, Sochen N, Cohen Y. Single- and double-Diffusion encoding MRI for studying ex vivo apparent axon diameter distribution in spinal cord white matter. NMR IN BIOMEDICINE 2019; 32:e4170. [PMID: 31573745 DOI: 10.1002/nbm.4170] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 07/28/2019] [Accepted: 07/31/2019] [Indexed: 06/10/2023]
Abstract
Mapping average axon diameter (AAD) and axon diameter distribution (ADD) in neuronal tissues non-invasively is a challenging task that may have a tremendous effect on our understanding of the normal and diseased central nervous system (CNS). Water diffusion is used to probe microstructure in neuronal tissues, however, the different water populations and barriers that are present in these tissues turn this into a complex task. Therefore, it is not surprising that recently we have witnessed a burst in the development of new approaches and models that attempt to obtain, non-invasively, detailed microstructural information in the CNS. In this work, we aim at challenging and comparing the microstructural information obtained from single diffusion encoding (SDE) with double diffusion encoding (DDE) MRI. We first applied SDE and DDE MR spectroscopy (MRS) on microcapillary phantoms and then applied SDE and DDE MRI on an ex vivo porcine spinal cord (SC), using similar experimental conditions. The obtained diffusion MRI data were fitted by the same theoretical model, assuming that the signal in every voxel can be approximated as the superposition of a Gaussian-diffusing component and a series of restricted components having infinite cylindrical geometries. The diffusion MRI results were then compared with histological findings. We found a good agreement between the fittings and the experimental data in white matter (WM) voxels of the SC in both diffusion MRI methods. The microstructural information and apparent AADs extracted from SDE MRI were found to be similar or somewhat larger than those extracted from DDE MRI especially when the diffusion time was set to 40 ms. The apparent ADDs extracted from SDE and DDE MRI show reasonable agreement but somewhat weaker correspondence was observed between the diffusion MRI results and histology. The apparent subtle differences between the microstructural information obtained from SDE and DDE MRI are briefly discussed.
Collapse
Affiliation(s)
- Debbie Anaby
- School of Chemistry, The Raymond and Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, Tel Aviv, Israel
- Department of Diagnostic Imaging, Sheba Medical Center, Tel HaShomer, Israel
| | - Darya Morozov
- School of Chemistry, The Raymond and Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Inbar Seroussi
- School of Mathematical Sciences, The Raymond and Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Simon Hametner
- Neuroimmunology Department, Center of Brain Research, Medical University of Vienna, Vienna, Austria
| | - Nir Sochen
- School of Mathematical Sciences, The Raymond and Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, Tel Aviv, Israel
- The Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Yoram Cohen
- School of Chemistry, The Raymond and Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, Tel Aviv, Israel
- The Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
20
|
Fritsche L, Teuber-Hanselmann S, Soub D, Harnisch K, Mairinger F, Junker A. MicroRNA profiles of MS gray matter lesions identify modulators of the synaptic protein synaptotagmin-7. Brain Pathol 2019; 30:524-540. [PMID: 31663645 PMCID: PMC8018161 DOI: 10.1111/bpa.12800] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 10/24/2019] [Indexed: 12/11/2022] Open
Abstract
We established microRNA (miRNA) profiles in gray and white matter multiple sclerosis (MS) lesions and identified seven miRNAs which were significantly more upregulated in the gray matter lesions. Five of those seven miRNAs, miR‐330‐3p, miR‐4286, miR‐4488, let‐7e‐5p, miR‐432‐5p shared the common target synaptotagmin7 (Syt7). Immunohistochemistry and transcript analyses using nanostring technology revealed a maldistribution of Syt7, with Syt7 accumulation in neuronal soma and decreased expression in axonal structures. This maldistribution could be at least partially explained by an axonal Syt7 transport disturbance. Since Syt7 is a synapse‐associated molecule, this maldistribution could result in impairment of neuronal functions in MS patients. Thus, our results lead to the hypothesis that the overexpression of these five miRNAs in gray matter lesions is a cellular mechanism to reduce further endogenous neuronal Syt7 production. Therefore, miRNAs seem to play an important role as modulators of neuronal structures in MS.
Collapse
Affiliation(s)
- Lena Fritsche
- Institute of Neuropathology, University Hospital Essen, D-45147, Essen, Germany
| | | | - Daniel Soub
- Institute of Neuropathology, University Hospital Essen, D-45147, Essen, Germany
| | - Kim Harnisch
- Institute of Neuropathology, University Hospital Essen, D-45147, Essen, Germany
| | - Fabian Mairinger
- Institute of Pathology, University Hospital Essen, D-45147, Essen, Germany
| | - Andreas Junker
- Institute of Neuropathology, University Hospital Essen, D-45147, Essen, Germany
| |
Collapse
|
21
|
Stadelmann C, Timmler S, Barrantes-Freer A, Simons M. Myelin in the Central Nervous System: Structure, Function, and Pathology. Physiol Rev 2019; 99:1381-1431. [PMID: 31066630 DOI: 10.1152/physrev.00031.2018] [Citation(s) in RCA: 325] [Impact Index Per Article: 65.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Oligodendrocytes generate multiple layers of myelin membrane around axons of the central nervous system to enable fast and efficient nerve conduction. Until recently, saltatory nerve conduction was considered the only purpose of myelin, but it is now clear that myelin has more functions. In fact, myelinating oligodendrocytes are embedded in a vast network of interconnected glial and neuronal cells, and increasing evidence supports an active role of oligodendrocytes within this assembly, for example, by providing metabolic support to neurons, by regulating ion and water homeostasis, and by adapting to activity-dependent neuronal signals. The molecular complexity governing these interactions requires an in-depth molecular understanding of how oligodendrocytes and axons interact and how they generate, maintain, and remodel their myelin sheaths. This review deals with the biology of myelin, the expanded relationship of myelin with its underlying axons and the neighboring cells, and its disturbances in various diseases such as multiple sclerosis, acute disseminated encephalomyelitis, and neuromyelitis optica spectrum disorders. Furthermore, we will highlight how specific interactions between astrocytes, oligodendrocytes, and microglia contribute to demyelination in hereditary white matter pathologies.
Collapse
Affiliation(s)
- Christine Stadelmann
- Institute of Neuropathology, University Medical Center Göttingen , Göttingen , Germany ; Institute of Neuronal Cell Biology, Technical University Munich , Munich , Germany ; German Center for Neurodegenerative Diseases (DZNE), Munich , Germany ; Department of Neuropathology, University Medical Center Leipzig , Leipzig , Germany ; Munich Cluster of Systems Neurology (SyNergy), Munich , Germany ; and Max Planck Institute of Experimental Medicine, Göttingen , Germany
| | - Sebastian Timmler
- Institute of Neuropathology, University Medical Center Göttingen , Göttingen , Germany ; Institute of Neuronal Cell Biology, Technical University Munich , Munich , Germany ; German Center for Neurodegenerative Diseases (DZNE), Munich , Germany ; Department of Neuropathology, University Medical Center Leipzig , Leipzig , Germany ; Munich Cluster of Systems Neurology (SyNergy), Munich , Germany ; and Max Planck Institute of Experimental Medicine, Göttingen , Germany
| | - Alonso Barrantes-Freer
- Institute of Neuropathology, University Medical Center Göttingen , Göttingen , Germany ; Institute of Neuronal Cell Biology, Technical University Munich , Munich , Germany ; German Center for Neurodegenerative Diseases (DZNE), Munich , Germany ; Department of Neuropathology, University Medical Center Leipzig , Leipzig , Germany ; Munich Cluster of Systems Neurology (SyNergy), Munich , Germany ; and Max Planck Institute of Experimental Medicine, Göttingen , Germany
| | - Mikael Simons
- Institute of Neuropathology, University Medical Center Göttingen , Göttingen , Germany ; Institute of Neuronal Cell Biology, Technical University Munich , Munich , Germany ; German Center for Neurodegenerative Diseases (DZNE), Munich , Germany ; Department of Neuropathology, University Medical Center Leipzig , Leipzig , Germany ; Munich Cluster of Systems Neurology (SyNergy), Munich , Germany ; and Max Planck Institute of Experimental Medicine, Göttingen , Germany
| |
Collapse
|
22
|
Singh V, Tripathi A, Dutta R. Proteomic Approaches to Decipher Mechanisms Underlying Pathogenesis in Multiple Sclerosis Patients. Proteomics 2019; 19:e1800335. [PMID: 31119864 PMCID: PMC6690771 DOI: 10.1002/pmic.201800335] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 05/15/2019] [Indexed: 12/13/2022]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory demyelinating and neurodegenerative disease of the central nervous system (CNS). The cause of MS is unknown, with no effective therapies available to halt the progressive neurological disability. Development of new and improvement of existing therapeutic strategies therefore require a better understanding of MS pathogenesis, especially during the progressive phase of the disease. This can be achieved through development of biomarkers that can help to identify disease pathophysiology and monitor disease progression. Proteomics is a powerful and promising tool to accelerate biomarker detection and contribute to novel therapeutics. In this review, an overview of how proteomic technology using CNS tissues and biofluids from MS patients has provided important clues to the pathogenesis of MS is provided. Current publications, pitfalls, as well as directions of future research involving proteomic approaches to understand the pathogenesis of MS are discussed.
Collapse
Affiliation(s)
- Vaibhav Singh
- Department of Neurosciences, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Ajai Tripathi
- Department of Neurosciences, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Ranjan Dutta
- Department of Neurosciences, Cleveland Clinic, Cleveland, OH, 44195, USA
| |
Collapse
|
23
|
Neuronal vulnerability and multilineage diversity in multiple sclerosis. Nature 2019; 573:75-82. [PMID: 31316211 PMCID: PMC6731122 DOI: 10.1038/s41586-019-1404-z] [Citation(s) in RCA: 328] [Impact Index Per Article: 65.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 06/12/2019] [Indexed: 02/06/2023]
Abstract
Multiple sclerosis (MS) is a neuroinflammatory disease with a relapsing-remitting disease course at early stages, distinct lesion characteristics in cortical gray versus subcortical white matter, and neurodegeneration at chronic stages. We assessed multilineage cell expression changes using single-nucleus RNA sequencing (snRNA-seq) and validated results using multiplex in situ hybridization in MS lesions. We found selective vulnerability and loss of excitatory CUX2-expressing projection neurons in upper cortical layers underlying meningeal inflammation; such MS neuron populations showed upregulation of stress pathway genes and long non-coding RNAs. Signatures of stressed oligodendrocytes, reactive astrocytes and activated phagocytosing cells mapped most strongly to the rim of MS plaques. Interestingly, snRNA-seq identified phagocytosing microglia and/or macrophages by their ingestion and perinuclear import of myelin transcripts, confirmed by functional mouse and human culture assays. Our findings indicate lineage- and region-specific transcriptomic changes associated with selective cortical neuron damage and glial activation contributing to MS lesion progression.
Collapse
|
24
|
Ciccarelli O, Cohen JA, Reingold SC, Weinshenker BG, Amato MP, Banwell B, Barkhof F, Bebo B, Becher B, Bethoux F, Brandt A, Brownlee W, Calabresi P, Chatway J, Chien C, Chitnis T, Ciccarelli O, Cohen J, Comi G, Correale J, De Sèze J, De Stefano N, Fazekas F, Flanagan E, Freedman M, Fujihara K, Galetta S, Goldman M, Greenberg B, Hartung HP, Hemmer B, Henning A, Izbudak I, Kappos L, Lassmann H, Laule C, Levy M, Lublin F, Lucchinetti C, Lukas C, Marrie RA, Miller A, Miller D, Montalban X, Mowry E, Ourselin S, Paul F, Pelletier D, Ranjeva JP, Reich D, Reingold S, Rocca MA, Rovira A, Schlaerger R, Soelberg Sorensen P, Sormani M, Stuve O, Thompson A, Tintoré M, Traboulsee A, Trapp B, Trojano M, Uitdehaag B, Vukusic S, Waubant E, Weinshenker B, Wheeler-Kingshott CG, Xu J. Spinal cord involvement in multiple sclerosis and neuromyelitis optica spectrum disorders. Lancet Neurol 2019; 18:185-197. [DOI: 10.1016/s1474-4422(18)30460-5] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 11/09/2018] [Accepted: 11/14/2018] [Indexed: 12/13/2022]
|
25
|
Gorter RP, Nutma E, Jahrei M, de Jonge JC, Quinlan RA, van der Valk P, van Noort JM, Baron W, Amor S. Heat shock proteins are differentially expressed in brain and spinal cord: implications for multiple sclerosis. Clin Exp Immunol 2018; 194:137-152. [PMID: 30014472 PMCID: PMC6194336 DOI: 10.1111/cei.13186] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/11/2018] [Indexed: 01/10/2023] Open
Abstract
Multiple sclerosis (MS) is a chronic neurodegenerative disease characterized by demyelination, inflammation and neurodegeneration throughout the central nervous system. Although spinal cord pathology is an important factor contributing to disease progression, few studies have examined MS lesions in the spinal cord and how they differ from brain lesions. In this study we have compared brain and spinal cord white (WM) and grey (GM) matter from MS and control tissues, focusing on small heat shock proteins (HSPB) and HSP16.2. Western blotting was used to examine protein levels of HSPB1, HSPB5, HSPB6, HSPB8 and HSP16.2 in brain and spinal cord from MS and age-matched non-neurological controls. Immunohistochemistry was used to examine expression of the HSPs in MS spinal cord lesions and controls. Expression levels were quantified using ImageJ. Western blotting revealed significantly higher levels of HSPB1, HSPB6 and HSPB8 in MS and control spinal cord compared to brain tissues. No differences in HSPB5 and HSP16.2 protein levels were observed, although HSPB5 protein levels were higher in brain WM versus GM. In MS spinal cord lesions, increased HSPB1 and HSPB5 expression was observed in astrocytes, and increased neuronal expression of HSP16.2 was observed in normal-appearing GM and type 1 GM lesions. The high constitutive expression of several HSPBs in spinal cord and increased expression of HSPBs and HSP16.2 in MS illustrate differences between brain and spinal cord in health and upon demyelination. Regional differences in HSP expression may reflect differences in astrocyte cytoskeleton composition and influence inflammation, possibly affecting the effectiveness of pharmacological agents.
Collapse
Affiliation(s)
- R. P. Gorter
- Pathology DepartmentAmsterdam UMC, VUMCGroningenUK
| | - E. Nutma
- Pathology DepartmentAmsterdam UMC, VUMCGroningenUK
| | - M.‐C. Jahrei
- Pathology DepartmentAmsterdam UMC, VUMCGroningenUK
| | - J. C. de Jonge
- Department of Cell BiologyUniversity of Groningen, University Medical Center GroningenGroningenUK
| | - R. A Quinlan
- Department of BiosciencesDurham UniversityDurhamUK
| | | | | | - W. Baron
- Department of Cell BiologyUniversity of Groningen, University Medical Center GroningenGroningenUK
| | - S. Amor
- Pathology DepartmentAmsterdam UMC, VUMCGroningenUK
- Centre for Neuroscience and Trauma, Blizard Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| |
Collapse
|
26
|
Tahedl M, Levine SM, Greenlee MW, Weissert R, Schwarzbach JV. Functional Connectivity in Multiple Sclerosis: Recent Findings and Future Directions. Front Neurol 2018; 9:828. [PMID: 30364281 PMCID: PMC6193088 DOI: 10.3389/fneur.2018.00828] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 09/14/2018] [Indexed: 02/03/2023] Open
Abstract
Multiple sclerosis is a debilitating disorder resulting from scattered lesions in the central nervous system. Because of the high variability of the lesion patterns between patients, it is difficult to relate existing biomarkers to symptoms and their progression. The scattered nature of lesions in multiple sclerosis offers itself to be studied through the lens of network analyses. Recent research into multiple sclerosis has taken such a network approach by making use of functional connectivity. In this review, we briefly introduce measures of functional connectivity and how to compute them. We then identify several common observations resulting from this approach: (a) high likelihood of altered connectivity in deep-gray matter regions, (b) decrease of brain modularity, (c) hemispheric asymmetries in connectivity alterations, and (d) correspondence of behavioral symptoms with task-related and task-unrelated networks. We propose incorporating such connectivity analyses into longitudinal studies in order to improve our understanding of the underlying mechanisms affected by multiple sclerosis, which can consequently offer a promising route to individualizing imaging-related biomarkers for multiple sclerosis.
Collapse
Affiliation(s)
- Marlene Tahedl
- Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg, Germany
- Institute for Experimental Psychology, University of Regensburg, Regensburg, Germany
| | - Seth M. Levine
- Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg, Germany
| | - Mark W. Greenlee
- Institute for Experimental Psychology, University of Regensburg, Regensburg, Germany
| | - Robert Weissert
- Department of Neurology, University of Regensburg, Regensburg, Germany
| | - Jens V. Schwarzbach
- Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg, Germany
| |
Collapse
|
27
|
Visualization of the Breakdown of the Axonal Transport Machinery: a Comparative Ultrastructural and Immunohistochemical Approach. Mol Neurobiol 2018; 56:3984-3998. [PMID: 30238390 DOI: 10.1007/s12035-018-1353-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 09/13/2018] [Indexed: 12/22/2022]
Abstract
Axonal damage is a major factor contributing to disease progression in multiple sclerosis (MS) patients. On the histological level, acute axonal injury is most frequently analyzed by anti-amyloid precursor protein immunohistochemistry. To what extent this method truly detects axonal injury, and whether other proteins and organelles are as well subjected to axonal transport deficits in demyelinated tissues is not known. The aim of this study was to correlate ultrastructural morphology with the immunohistochemical appearance of acute axonal injury in a model of toxin-induced oligodendrocyte degeneration. C57BL/6J mice were intoxicated with 0.25% cuprizone to induce demyelination. The corpus callosum was investigated by serial block-face scanning electron microscopy (i.e., 3D EM), immunohistochemistry, and immunofluorescence microscopy. Brain tissues of progressive MS patients were included to test the relevance of our findings in mice for MS. Volumes of axonal swellings, determined by 3D EM, were comparable to volumes of axonal spheroids, determined by anti-APP immunofluorescence stains. Axonal swellings were present at myelinated and non-myelinated axonal internodes. Densities of amyloid precursor protein (APP)+ spheroids were highest during active demyelination. Besides APP, vesicular glutamate transporter 1 and mitochondrial proteins accumulated at sites of axonal spheroids. Such accumulations were found as well in lesions of progressive MS patients. In this correlative ultrastructural-immunohistochemical study, we provide strong evidence that breakdown of the axonal transport machinery results in focal accumulations of mitochondria and different synaptic proteins. We provide new marker proteins to visualize acute axonal injury, which helps to further understand the complex nature of axonal damage in progressive MS.
Collapse
|
28
|
Schirmer L, Möbius W, Zhao C, Cruz-Herranz A, Ben Haim L, Cordano C, Shiow LR, Kelley KW, Sadowski B, Timmons G, Pröbstel AK, Wright JN, Sin JH, Devereux M, Morrison DE, Chang SM, Sabeur K, Green AJ, Nave KA, Franklin RJ, Rowitch DH. Oligodendrocyte-encoded Kir4.1 function is required for axonal integrity. eLife 2018; 7:36428. [PMID: 30204081 PMCID: PMC6167053 DOI: 10.7554/elife.36428] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 09/09/2018] [Indexed: 12/17/2022] Open
Abstract
Glial support is critical for normal axon function and can become dysregulated in white matter (WM) disease. In humans, loss-of-function mutations of KCNJ10, which encodes the inward-rectifying potassium channel KIR4.1, causes seizures and progressive neurological decline. We investigated Kir4.1 functions in oligodendrocytes (OLs) during development, adulthood and after WM injury. We observed that Kir4.1 channels localized to perinodal areas and the inner myelin tongue, suggesting roles in juxta-axonal K+ removal. Conditional knockout (cKO) of OL-Kcnj10 resulted in late onset mitochondrial damage and axonal degeneration. This was accompanied by neuronal loss and neuro-axonal dysfunction in adult OL-Kcnj10 cKO mice as shown by delayed visual evoked potentials, inner retinal thinning and progressive motor deficits. Axon pathologies in OL-Kcnj10 cKO were exacerbated after WM injury in the spinal cord. Our findings point towards a critical role of OL-Kir4.1 for long-term maintenance of axonal function and integrity during adulthood and after WM injury.
Collapse
Affiliation(s)
- Lucas Schirmer
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, California, United States.,Department of Pediatrics, University of California, San Francisco, San Francisco, United States.,Department of Paediatrics, University of Cambridge, Cambridge, United Kingdom.,Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Wiebke Möbius
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany.,Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany
| | - Chao Zhao
- Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom.,Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Andrés Cruz-Herranz
- Department of Neurology, University of California, San Francisco, San Francisco, United States.,Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, United States
| | - Lucile Ben Haim
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, California, United States.,Department of Pediatrics, University of California, San Francisco, San Francisco, United States
| | - Christian Cordano
- Department of Neurology, University of California, San Francisco, San Francisco, United States.,Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, United States
| | - Lawrence R Shiow
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, California, United States.,Department of Pediatrics, University of California, San Francisco, San Francisco, United States
| | - Kevin W Kelley
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, California, United States.,Department of Pediatrics, University of California, San Francisco, San Francisco, United States
| | - Boguslawa Sadowski
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany.,Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany
| | - Garrett Timmons
- Department of Neurology, University of California, San Francisco, San Francisco, United States.,Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, United States
| | - Anne-Katrin Pröbstel
- Department of Neurology, University of California, San Francisco, San Francisco, United States.,Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, United States
| | - Jackie N Wright
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, California, United States.,Department of Pediatrics, University of California, San Francisco, San Francisco, United States
| | - Jung Hyung Sin
- Department of Neurology, University of California, San Francisco, San Francisco, United States.,Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, United States
| | - Michael Devereux
- Department of Neurology, University of California, San Francisco, San Francisco, United States.,Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, United States
| | - Daniel E Morrison
- Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom.,Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Sandra M Chang
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, California, United States.,Department of Pediatrics, University of California, San Francisco, San Francisco, United States
| | - Khalida Sabeur
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, California, United States.,Department of Pediatrics, University of California, San Francisco, San Francisco, United States
| | - Ari J Green
- Department of Neurology, University of California, San Francisco, San Francisco, United States.,Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, United States.,Department of Ophthalmology, University of California, San Francisco, San Francisco, United States
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany.,Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany
| | - Robin Jm Franklin
- Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom.,Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - David H Rowitch
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, California, United States.,Department of Pediatrics, University of California, San Francisco, San Francisco, United States.,Department of Paediatrics, University of Cambridge, Cambridge, United Kingdom.,Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom.,Department of Neurosurgery, University of California, San Francisco, San Francisco, United States
| |
Collapse
|
29
|
Ingwersen J, De Santi L, Wingerath B, Graf J, Koop B, Schneider R, Hecker C, Schröter F, Bayer M, Engelke AD, Dietrich M, Albrecht P, Hartung HP, Annunziata P, Aktas O, Prozorovski T. Nimodipine confers clinical improvement in two models of experimental autoimmune encephalomyelitis. J Neurochem 2018; 146:86-98. [PMID: 29473171 DOI: 10.1111/jnc.14324] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 02/07/2018] [Accepted: 02/08/2018] [Indexed: 12/31/2022]
Abstract
Multiple sclerosis is characterised by inflammatory neurodegeneration, with axonal injury and neuronal cell death occurring in parallel to demyelination. Regarding the molecular mechanisms responsible for demyelination and axonopathy, energy failure, aberrant expression of ion channels and excitotoxicity have been suggested to lead to Ca2+ overload and subsequent activation of calcium-dependent damage pathways. Thus, the inhibition of Ca2+ influx by pharmacological modulation of Ca2+ channels may represent a novel neuroprotective strategy in the treatment of secondary axonopathy. We therefore investigated the effects of the L-type voltage-gated calcium channel blocker nimodipine in two different models of mouse experimental autoimmune encephalomyelitis (EAE), an established experimental paradigm for multiple sclerosis. We show that preventive application of nimodipine (10 mg/kg per day) starting on the day of induction had ameliorating effects on EAE in SJL/J mice immunised with encephalitic myelin peptide PLP139-151 , specifically in late-stage disease. Furthermore, supporting these data, administration of nimodipine to MOG35-55 -immunised C57BL/6 mice starting at the peak of pre-established disease, also led to a significant decrease in disease score, indicating a protective effect on secondary CNS damage. Histological analysis confirmed that nimodipine attenuated demyelination, axonal loss and pathological axonal β-amyloid precursor protein accumulation in the cerebellum and spinal cord in the chronic phase of disease. Of note, we observed no effects of nimodipine on the peripheral immune response in EAE mice with regard to distribution, antigen-specific proliferation or activation patterns of lymphocytes. Taken together, our data suggest a CNS-specific effect of L-type voltage-gated calcium channel blockade to inflammation-induced neurodegeneration.
Collapse
Affiliation(s)
- Jens Ingwersen
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Lorenzo De Santi
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Britta Wingerath
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Jonas Graf
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Barbara Koop
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Reiner Schneider
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Christina Hecker
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Friederike Schröter
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Mary Bayer
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Anna Dorothee Engelke
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Michael Dietrich
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Philipp Albrecht
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Hans-Peter Hartung
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Pasquale Annunziata
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Orhan Aktas
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Tim Prozorovski
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| |
Collapse
|
30
|
Stojic A, Bojcevski J, Williams SK, Diem R, Fairless R. Early Nodal and Paranodal Disruption in Autoimmune Optic Neuritis. J Neuropathol Exp Neurol 2018; 77:361-373. [DOI: 10.1093/jnen/nly011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Aleksandar Stojic
- Department of Neurology, University Clinic Heidelberg, Heidelberg, Germany
| | - Jovana Bojcevski
- Department of Neurology, University Clinic Heidelberg, Heidelberg, Germany
| | - Sarah K Williams
- Department of Neurology, University Clinic Heidelberg, Heidelberg, Germany
| | - Ricarda Diem
- Department of Neurology, University Clinic Heidelberg, Heidelberg, Germany
| | - Richard Fairless
- Department of Neurology, University Clinic Heidelberg, Heidelberg, Germany
| |
Collapse
|
31
|
Hartlehnert M, Derksen A, Hagenacker T, Kindermann D, Schäfers M, Pawlak M, Kieseier BC, Meyer Zu Horste G. Schwann cells promote post-traumatic nerve inflammation and neuropathic pain through MHC class II. Sci Rep 2017; 7:12518. [PMID: 28970572 PMCID: PMC5624882 DOI: 10.1038/s41598-017-12744-2] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 09/15/2017] [Indexed: 12/20/2022] Open
Abstract
The activation of T helper cells requires antigens to be exposed on the surface of antigen presenting cells (APCs) via MHC class II (MHC-II) molecules. Expression of MHC-II is generally limited to professional APCs, but other cell types can express MHC-II under inflammatory conditions. However, the importance of these conditional APCs is unknown. We and others have previously shown that Schwann cells are potentially conditional APCs, but the functional relevance of MHC-II expression by Schwann cells has not been studied in vivo. Here, we conditionally deleted the MHC-II β-chain from myelinating Schwann cells in mice and investigated how this influenced post-traumatic intraneural inflammation and neuropathic pain using the chronic constriction injury (CCI) model. We demonstrate that deletion of MHC-II in myelinating Schwann cells reduces thermal hyperalgesia and, to a lesser extent, also diminishes mechanical allodynia in CCI in female mice. This was accompanied by a reduction of intraneural CD4+ T cells and greater preservation of preferentially large-caliber axons. Activation of T helper cells by MHC-II on Schwann cells thus promotes post-traumatic axonal loss and neuropathic pain. Hence, we provide experimental evidence that Schwann cells gain antigen-presenting function in vivo and modulate local immune responses and diseases in the peripheral nerves.
Collapse
Affiliation(s)
- Maike Hartlehnert
- Department of Neurology, University Hospital Münster, Münster, Germany
| | - Angelika Derksen
- Department of Neurology, Heinrich-Heine-University, Medical Faculty, Düsseldorf, Germany
| | - Tim Hagenacker
- Department of Neurology, University of Duisburg-Essen, Essen, Germany
| | - David Kindermann
- Department of Neurology, University of Duisburg-Essen, Essen, Germany
| | - Maria Schäfers
- Department of Neurology, University of Duisburg-Essen, Essen, Germany
| | - Mathias Pawlak
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
| | - Bernd C Kieseier
- Department of Neurology, Heinrich-Heine-University, Medical Faculty, Düsseldorf, Germany
| | - Gerd Meyer Zu Horste
- Department of Neurology, University Hospital Münster, Münster, Germany. .,Department of Neurology, Heinrich-Heine-University, Medical Faculty, Düsseldorf, Germany.
| |
Collapse
|
32
|
Zhou T, Ahmad TK, Gozda K, Truong J, Kong J, Namaka M. Implications of white matter damage in amyotrophic lateral sclerosis (Review). Mol Med Rep 2017; 16:4379-4392. [PMID: 28791401 PMCID: PMC5646997 DOI: 10.3892/mmr.2017.7186] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2017] [Accepted: 06/09/2017] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease, which involves the progressive degeneration of motor neurons. ALS has long been considered a disease of the grey matter; however, pathological alterations of the white matter (WM), including axonal loss, axonal demyelination and oligodendrocyte death, have been reported in patients with ALS. The present review examined motor neuron death as the primary cause of ALS and evaluated the associated WM damage that is guided by neuronal‑glial interactions. Previous studies have suggested that WM damage may occur prior to the death of motor neurons, and thus may be considered an early indicator for the diagnosis and prognosis of ALS. However, the exact molecular mechanisms underlying early‑onset WM damage in ALS have yet to be elucidated. The present review explored the detailed anatomy of WM and identified several pathological mechanisms that may be implicated in WM damage in ALS. In addition, it associated the pathophysiological alterations of WM, which may contribute to motor neuron death in ALS, with similar mechanisms of WM damage that are involved in multiple sclerosis (MS). Furthermore, the early detection of WM damage in ALS, using neuroimaging techniques, may lead to earlier therapeutic intervention, using immunomodulatory treatment strategies similar to those used in relapsing‑remitting MS, aimed at delaying WM damage in ALS. Early therapeutic approaches may have the potential to delay motor neuron damage and thus prolong the survival of patients with ALS. The therapeutic interventions that are currently available for ALS are only marginally effective. However, early intervention with immunomodulatory drugs may slow the progression of WM damage in the early stages of ALS, thus delaying motor neuron death and increasing the life expectancy of patients with ALS.
Collapse
Affiliation(s)
- Ting Zhou
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
- Department of Human Anatomy and Cell Science, College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Tina Khorshid Ahmad
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
| | - Kiana Gozda
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
| | - Jessica Truong
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
| | - Jiming Kong
- Department of Human Anatomy and Cell Science, College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Michael Namaka
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
- Department of Human Anatomy and Cell Science, College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
- College of Pharmacy, Third Military Medical University, Chongqing 400038, P.R. China
- Department of Medical Rehabilitation, College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0T6, Canada
- Department of Internal Medicine, College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 1R9, Canada
| |
Collapse
|
33
|
Ohgomori T, Yamasaki R, Takeuchi H, Kadomatsu K, Kira JI, Jinno S. Differential activation of neuronal and glial STAT3 in the spinal cord of the SOD1 G93A mouse model of amyotrophic lateral sclerosis. Eur J Neurosci 2017; 46:2001-2014. [PMID: 28715117 DOI: 10.1111/ejn.13650] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 07/05/2017] [Accepted: 07/10/2017] [Indexed: 12/14/2022]
Abstract
Signal transducer and activator of transcription (STAT) proteins are activated by phosphorylation in the spinal cord of patients suffering from amyotrophic lateral sclerosis (ALS). The major scope of our study is a comprehensive histological characterization of the mechanisms underlying neuronal and glial STAT3 activation in the pathogenesis of ALS using SOD1G93A mice. We calculated the fold changes (FCs, ratios vs. appropriate controls) of the numerical densities of the following phosphorylated STAT3-positive (pSTAT3)+ cells - choline acetyltransferase (ChAT)+ α-motoneurons, ionized calcium-binding adapter molecule 1 (Iba1)+ microglia, and S100β+ astrocytes in SOD1G93A mice. The FCs of pSTAT3+ microglia and pSTAT3+ astrocytes were increased from 9 to 15 weeks of age and then plateaued until 21 weeks. In contrast, the FCs of pSTAT3+ α-motoneurons peaked at 9 weeks and then decreased until 21 weeks. The immunoreactivity for nonphosphorylated neurofilament protein (SMI-32), a marker of axonal impairment, was decreased in pSTAT3+ α-motoneurons compared with pSTAT3- α-motoneurons at 9 weeks of age. We then compared the following pharmacological models - the chronic administration of 3,3'-iminodipropionitrile (IDPN), which models axonal impairment, and the acute administration of lipopolysaccharide (LPS), which is a model of neuroinflammation. The FCs of pSTAT3+ α-motoneurons were increased in IDPN-treated mice, while those of pSTAT3+ microglia were increased in LPS-treated mice. The FCs of pSTAT3+ astrocytes were higher in SOD1G93A mice at 9 weeks compared with IDPN- and LPS-treated mice. Our results indicate that axonopathy and neuroinflammation may trigger the respective activation of neuronal and glial STAT3, which is observed during ALS pathogenesis.
Collapse
Affiliation(s)
- Tomohiro Ohgomori
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Ryo Yamasaki
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hideyuki Takeuchi
- Department of Neuroimmunology, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Kenji Kadomatsu
- Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Jun-Ichi Kira
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shozo Jinno
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| |
Collapse
|
34
|
Lagumersindez-Denis N, Wrzos C, Mack M, Winkler A, van der Meer F, Reinert MC, Hollasch H, Flach A, Brühl H, Cullen E, Schlumbohm C, Fuchs E, Linington C, Barrantes-Freer A, Metz I, Wegner C, Liebetanz D, Prinz M, Brück W, Stadelmann C, Nessler S. Differential contribution of immune effector mechanisms to cortical demyelination in multiple sclerosis. Acta Neuropathol 2017; 134:15-34. [PMID: 28386765 PMCID: PMC5486638 DOI: 10.1007/s00401-017-1706-x] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 03/08/2017] [Accepted: 03/25/2017] [Indexed: 12/16/2022]
Abstract
Cortical demyelination is a widely recognized hallmark of multiple sclerosis (MS) and correlate of disease progression and cognitive decline. The pathomechanisms initiating and driving gray matter damage are only incompletely understood. Here, we determined the infiltrating leukocyte subpopulations in 26 cortical demyelinated lesions of biopsied MS patients and assessed their contribution to cortical lesion formation in a newly developed mouse model. We find that conformation-specific anti-myelin antibodies contribute to cortical demyelination even in the absence of the classical complement pathway. T cells and natural killer cells are relevant for intracortical type 2 but dispensable for subpial type 3 lesions, whereas CCR2+ monocytes are required for both. Depleting CCR2+ monocytes in marmoset monkeys with experimental autoimmune encephalomyelitis using a novel humanized CCR2 targeting antibody translates into significantly less cortical demyelination and disease severity. We conclude that biologics depleting CCR2+ monocytes might be attractive candidates for preventing cortical lesion formation and ameliorating disease progression in MS.
Collapse
|
35
|
Wood ET, Ercan E, Sati P, Cortese ICM, Ronen I, Reich DS. Longitudinal MR spectroscopy of neurodegeneration in multiple sclerosis with diffusion of the intra-axonal constituent N-acetylaspartate. Neuroimage Clin 2017; 15:780-788. [PMID: 28702353 PMCID: PMC5496488 DOI: 10.1016/j.nicl.2017.06.028] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 06/08/2017] [Accepted: 06/20/2017] [Indexed: 11/19/2022]
Abstract
Multiple sclerosis (MS) is a pathologically complex CNS disease: inflammation, demyelination, and neuroaxonal degeneration occur concurrently and may depend on one another. Current therapies are aimed at the immune-mediated, inflammatory destruction of myelin, whereas axonal degeneration is ongoing and not specifically targeted. Diffusion-weighted magnetic resonance spectroscopy can measure the diffusivity of metabolites in vivo, such as the axonal/neuronal constituent N-acetylaspartate, allowing compartment-specific assessment of disease-related changes. Previously, we found significantly lower N-acetylaspartate diffusivity in people with MS compared to healthy controls (Wood et al., 2012) suggesting that this technique can measure axonal degeneration and could be useful in developing neuroprotective agents. In this longitudinal study, we found that N-acetylaspartate diffusivity decreased by 8.3% (p < 0.05) over 6 months in participants who were experiencing clinical or MRI evidence of inflammatory activity (n = 13), whereas there was no significant change in N-acetylaspartate diffusivity in the context of clinical and radiological stability (n = 6). As N-acetylaspartate diffusivity measurements are thought to more specifically reflect the intra-axonal space, these data suggest that N-acetylaspartate diffusivity can report on axonal health on the background of multiple pathological processes in MS, both cross-sectionally and longitudinally.
Collapse
Key Words
- Axonopathy
- DW-MRS, diffusion-weighted magnetic resonance spectroscopy
- Diffusion-weighted magnetic resonance spectroscopy
- EDSS, Expanded Disability Scale Score
- HV, healthy volunteer
- ICV, intracranial volume
- MS, multiple sclerosis
- Multiple sclerosis
- NAA, N-acetylaspartate
- PASAT, Paced Auditory Symbol Addition Test
- T, Tesla
- VOI, volume of interest
- WM, white matter
- White matter
Collapse
Affiliation(s)
- Emily Turner Wood
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Ece Ercan
- C.J. Gorter Center for High Field MRI, Department of Radiology, Leiden University Medical Center, Leiden, Netherlands
| | - Pascal Sati
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Irene C M Cortese
- Neuroimmunology Clinic, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Itamar Ronen
- C.J. Gorter Center for High Field MRI, Department of Radiology, Leiden University Medical Center, Leiden, Netherlands
| | - Daniel S Reich
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA; Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
36
|
Gannon PJ, Akay-Espinoza C, Yee AC, Briand LA, Erickson MA, Gelman BB, Gao Y, Haughey NJ, Zink MC, Clements JE, Kim NS, Van De Walle G, Jensen BK, Vassar R, Pierce RC, Gill AJ, Kolson DL, Diehl JA, Mankowski JL, Jordan-Sciutto KL. HIV Protease Inhibitors Alter Amyloid Precursor Protein Processing via β-Site Amyloid Precursor Protein Cleaving Enzyme-1 Translational Up-Regulation. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:91-109. [PMID: 27993242 DOI: 10.1016/j.ajpath.2016.09.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 08/22/2016] [Accepted: 09/12/2016] [Indexed: 12/14/2022]
Abstract
Mounting evidence implicates antiretroviral (ARV) drugs as potential contributors to the persistence and evolution of clinical and pathological presentation of HIV-associated neurocognitive disorders in the post-ARV era. Based on their ability to induce endoplasmic reticulum (ER) stress in various cell types, we hypothesized that ARV-mediated ER stress in the central nervous system resulted in chronic dysregulation of the unfolded protein response and altered amyloid precursor protein (APP) processing. We used in vitro and in vivo models to show that HIV protease inhibitor (PI) class ARVs induced neuronal damage and ER stress, leading to PKR-like ER kinase-dependent phosphorylation of the eukaryotic translation initiation factor 2α and enhanced translation of β-site APP cleaving enzyme-1 (BACE1). In addition, PIs induced β-amyloid production, indicative of increased BACE1-mediated APP processing, in rodent neuroglial cultures and human APP-expressing Chinese hamster ovary cells. Inhibition of BACE1 activity protected against neuronal damage. Finally, ARVs administered to mice and SIV-infected macaques resulted in neuronal damage and BACE1 up-regulation in the central nervous system. These findings implicate a subset of PIs as potential mediators of neurodegeneration in HIV-associated neurocognitive disorders.
Collapse
Affiliation(s)
- Patrick J Gannon
- Department of Pharmacology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Pathology, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Cagla Akay-Espinoza
- Department of Pathology, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Alan C Yee
- Department of Pathology, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Lisa A Briand
- Department of Psychology, Temple University, Philadelphia, Pennsylvania
| | - Michelle A Erickson
- Department of Pathology, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Benjamin B Gelman
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas
| | - Yan Gao
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| | - Norman J Haughey
- Richard T. Johnson Division of Neuroimmunology and Neurological Infections, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - M Christine Zink
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Janice E Clements
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Nicholas S Kim
- Department of Pathology, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Gabriel Van De Walle
- Department of Pathology, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Brigid K Jensen
- Department of Pathology, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Robert Vassar
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - R Christopher Pierce
- Department of Psychiatry, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Alexander J Gill
- Department of Neurology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Dennis L Kolson
- Department of Neurology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - J Alan Diehl
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| | - Joseph L Mankowski
- Richard T. Johnson Division of Neuroimmunology and Neurological Infections, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Kelly L Jordan-Sciutto
- Department of Pathology, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
37
|
O'Sullivan SA, Velasco-Estevez M, Dev KK. Demyelination induced by oxidative stress is regulated by sphingosine 1-phosphate receptors. Glia 2017; 65:1119-1136. [PMID: 28375547 DOI: 10.1002/glia.23148] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 03/02/2017] [Accepted: 03/20/2017] [Indexed: 01/14/2023]
Abstract
Oxidative stress is a pathological condition defined as an imbalance between production and removal of reactive oxygen species. This process causes structural cell damage, disrupts DNA repair and induces mitochondrial dysfunction. Many in vitro studies have used direct bolus application of H2 O2 to investigate the role of oxidative stress in cell culture. In this study, using mouse organotypic cerebellar slice cultures, the effects of H2 O2 -induced oxidative stress on myelination state were examined, using bolus concentrations of H2 O2 (0.1-1 mM) and low-continuous H2 O2 (∼20 μM) generated from glucose oxidase and catalase (GOX-CAT). Using these models, the potential therapeutic effects of pFTY720, an oral therapy used in multiple sclerosis, was also examined. We found bolus treatment of H2 O2 (0.5 mM) and, for the first time, low-continuous H2 O2 (GOX-CAT) to induce demyelination in organotypic slices. Both bolus H2 O2 and GOX-CAT treatments significantly decreased vimentin expression in these slice cultures as well as increased cell death in isolated astrocyte cultures. Importantly, pre-treatment with pFTY720 significantly attenuated both bolus H2 O2 and GOX-CAT-induced demyelination and the GOX-CAT-induced decrease in vimentin in cerebellar slices, without altering levels of the proinflammatory cytokines such as IL-6 and CX3CL1. We also observed increased SMI-32 immunoreactivity in the white matter tract induced by GOX-CAT indicating axonal damage, which was remarkably attenuated by pFTY720. Taken together, this data establishes a novel GOX-CAT model of demyelination and demonstrates that pFTY720 can act independently of inflammatory cytokines to attenuate decreases in vimentin, as well as axonal damage and demyelination induced by oxidative stress.
Collapse
Affiliation(s)
- Sinead A O'Sullivan
- Drug Development, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | | | - Kumlesh K Dev
- Drug Development, School of Medicine, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
38
|
ALS-linked FUS exerts a gain of toxic function involving aberrant p38 MAPK activation. Sci Rep 2017; 7:115. [PMID: 28273913 PMCID: PMC5428330 DOI: 10.1038/s41598-017-00091-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 01/30/2017] [Indexed: 12/14/2022] Open
Abstract
Mutations in Fused in Sarcoma/Translocated in Liposarcoma (FUS) cause familial forms of amyotrophic lateral sclerosis (ALS), a neurodegenerative disease characterized by progressive axonal degeneration mainly affecting motor neurons. Evidence from transgenic mouse models suggests mutant forms of FUS exert an unknown gain-of-toxic function in motor neurons, but mechanisms underlying this effect remain unknown. Towards this end, we studied the effect of wild type FUS (FUS WT) and three ALS-linked variants (G230C, R521G and R495X) on fast axonal transport (FAT), a cellular process critical for appropriate maintenance of axonal connectivity. All ALS-FUS variants impaired anterograde and retrograde FAT in squid axoplasm, whereas FUS WT had no effect. Misfolding of mutant FUS is implicated in this process, as the molecular chaperone Hsp110 mitigated these toxic effects. Interestingly, mutant FUS-induced impairment of FAT in squid axoplasm and of axonal outgrowth in mammalian primary motor neurons involved aberrant activation of the p38 MAPK pathway, as also reported for ALS-linked forms of Cu, Zn superoxide dismutase (SOD1). Accordingly, increased levels of active p38 MAPK were detected in post-mortem human ALS-FUS brain tissues. These data provide evidence for a novel gain-of-toxic function for ALS-linked FUS involving p38 MAPK activation.
Collapse
|
39
|
Hollingsworth E, Khouri J, Imitola J. Endogenous repair and development inspired therapy of neurodegeneration in progressive multiple sclerosis. Expert Rev Neurother 2017; 17:611-629. [DOI: 10.1080/14737175.2017.1287564] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Affiliation(s)
- Ethan Hollingsworth
- Laboratory for Neural Stem Cells and Functional Neurogenetics, University Wexner Medical Center, Biomedical Research Tower, Columbus, OH, USA
- Division of Neuroimmunology and Multiple Sclerosis and Departments of Neurology and Neuroscience. The Ohio State, University Wexner Medical Center, Biomedical Research Tower, Columbus, OH, USA
| | - Jamil Khouri
- Laboratory for Neural Stem Cells and Functional Neurogenetics, University Wexner Medical Center, Biomedical Research Tower, Columbus, OH, USA
- Division of Neuroimmunology and Multiple Sclerosis and Departments of Neurology and Neuroscience. The Ohio State, University Wexner Medical Center, Biomedical Research Tower, Columbus, OH, USA
| | - Jaime Imitola
- Laboratory for Neural Stem Cells and Functional Neurogenetics, University Wexner Medical Center, Biomedical Research Tower, Columbus, OH, USA
- Division of Neuroimmunology and Multiple Sclerosis and Departments of Neurology and Neuroscience. The Ohio State, University Wexner Medical Center, Biomedical Research Tower, Columbus, OH, USA
| |
Collapse
|
40
|
Gudi V, Gai L, Herder V, Tejedor LS, Kipp M, Amor S, Sühs KW, Hansmann F, Beineke A, Baumgärtner W, Stangel M, Skripuletz T. Synaptophysin Is a Reliable Marker for Axonal Damage. J Neuropathol Exp Neurol 2017; 76:109-125. [PMID: 28177496 DOI: 10.1093/jnen/nlw114] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Indexed: 11/13/2022] Open
Abstract
Synaptophysin is an abundant membrane protein of synaptic vesicles. The objective of this study was to determine the utility of identifying synaptophysin accumulations (spheroids/ovoids/bulbs) in CNS white matter as an immunohistochemical marker of axonal damage in demyelinating and neuroinflammatory conditions. We studied the cuprizone toxicity and Theiler’s murine encephalomyelitis virus (TMEV) infection models of demyelination and analyzed CNS tissue from patients with multiple sclerosis (MS). Synaptophysin colocalized with the amyloid precursor protein (APP), a well-known marker of axonal damage. In the cuprizone model, numerous pathological synaptophysin/APP-positive spheroids/ovoids were identified in the corpus callosum at the onset of demyelination; the extent of synaptophysin/APP-positive vesicle aggregates correlated with identified reactive microglia; during late and chronic demyelination, the majority of synaptophysin/APP-positive spheroids/ovoids resolved but a few remained, indicating persistent axonal damage; in the remyelination phase, scattered large synaptophysin/APP-positive bulbs persisted. In the TMEV model, only a few large- to medium-sized synaptophysin/APP-positive bulbs were found in demyelinated areas. In MS patient tissue samples, the bulbs appeared exclusively at the inflammatory edges of lesions. In conclusion, our data suggest that synaptophysin as a reliable marker of axonal damage in the CNS in inflammatory/demyelinating conditions.
Collapse
Affiliation(s)
- Viktoria Gudi
- Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Lijie Gai
- Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Vanessa Herder
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany.,Center for Systems Neuroscience, Hannover, Germany
| | - Laura Salinas Tejedor
- Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Hannover Medical School, Hannover, Germany.,Center for Systems Neuroscience, Hannover, Germany
| | - Markus Kipp
- Department of Anatomy II, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Sandra Amor
- Department of Pathology, VU University Medical Center, Amsterdam, The Netherlands
| | - Kurt-Wolfram Sühs
- Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Florian Hansmann
- Center for Systems Neuroscience, Hannover, Germany.,Department of Pathology, VU University Medical Center, Amsterdam, The Netherlands
| | - Andreas Beineke
- Center for Systems Neuroscience, Hannover, Germany.,Department of Pathology, VU University Medical Center, Amsterdam, The Netherlands
| | - Wolfgang Baumgärtner
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Martin Stangel
- Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Hannover Medical School, Hannover, Germany.,Center for Systems Neuroscience, Hannover, Germany
| | - Thomas Skripuletz
- Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
41
|
Kawachi I, Lassmann H. Neurodegeneration in multiple sclerosis and neuromyelitis optica. J Neurol Neurosurg Psychiatry 2017; 88:137-145. [PMID: 27671902 DOI: 10.1136/jnnp-2016-313300] [Citation(s) in RCA: 188] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 09/05/2016] [Accepted: 09/07/2016] [Indexed: 11/04/2022]
Abstract
Multiple sclerosis (MS) and neuromyelitis optica (NMO) are autoimmune demyelinating diseases of the central nervous system (CNS), having distinct immunological and pathological features. They have two pathogenic components, 'inflammation' and 'neurodegeneration', with different degrees of severity and pathogenetic mechanisms. The target antigen of autoimmunity in NMO is the water channel aquaporin-4 (AQP4), and antibodies directed against this antigen result in astrocyte damage. MS is a disease primarily affecting myelin and oligodendrocytes, but thus far, no MS-specific autoantigen has been identified. The distinct inflammatory processes in these diseases may trigger cascades of events leading to disease-specific neurodegeneration. Damage of the CNS tissue appears to be amplified by mechanisms that are in part shared by the two conditions and involve oxidative burst activation in microglia/macrophages, mitochondrial damage and axonal energy failure, Wallerian degeneration and meningeal inflammation. However, they appear to differ regarding the nature of the inflammatory response, the type and extent of cortical injury, and the type of astrocyte reaction and damage. Here, we provide a detailed comparison of the pathology between MS and NMO, which may help to define shared and disease-specific mechanisms of neurodegeneration in these diseases.
Collapse
Affiliation(s)
- Izumi Kawachi
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Hans Lassmann
- Center for Brain Research, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
42
|
Kostic M, Zivkovic N, Cvetanovic A, Stojanovic I, Colic M. IL-17 signalling in astrocytes promotes glutamate excitotoxicity: Indications for the link between inflammatory and neurodegenerative events in multiple sclerosis. Mult Scler Relat Disord 2016; 11:12-17. [PMID: 28104249 DOI: 10.1016/j.msard.2016.11.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 10/24/2016] [Accepted: 11/13/2016] [Indexed: 11/15/2022]
Abstract
OBJECTIVE Th-17 cells have been exclusively referred to inflammatory events in multiple sclerosis (MS), while their importance in the development of glutamate excitotoxicity and the consequent neurodegeneration has been a completely unexplored concept. Accordingly, the objective of our study was to assess IL-17A effect on astrocyte ability to metabolize and release glutamate, considering that astrocytes had the central role in glutamate homeostasis. METHODS By using primary rat astrocyte cultures, astrocyte ability to uptake glutamate was estimated by the alterations of glutamate transporters (GLAST and GLT-1) expression, whereas changes in glutamine synthetase expression were used to estimate the ability to metabolize glutamate. Gene expression was determined by real time polymerase chain reaction (rtPCR). IL-17A effect on astrocyte ability to produce glutamate was investigated directly, by measuring the level of released glutamate using high performance liquid chromatography (HPLC). RESULTS Lower concentrations of IL-17A reduced the expressions of both glutamate transporters and glutamine synthetase; however, this effect was lost when IL-17A was applied in a higher dose. IL-17A did not significantly modify glutamate release from astrocyte in basal conditions, but following Ca2+ stimulation, as well as Ca2+ removal from the culture medium, IL-17A stimulated glutamate release in dose-dependent manner. CONCLUSION Together, these results support that IL-17A could promote glutamate excitotoxicity by decreasing astrocyte ability to uptake and convert glutamate to non-toxic glutamine, but also by stimulating Ca2+ dependent glutamate release. Such interactions between IL-17A and glutamate excitotoxicity implicate the potential link between inflammation and neurodegeneration during MS pathogenesis, and identify astrocytes as a potential target in achieving neuroprotective effects in MS.
Collapse
Affiliation(s)
- Milos Kostic
- Department of Immunology, Medical Faculty, University of Nis, Blvd. dr Zorana Djindjica 81, 18000 Nis, Serbia.
| | - Nikola Zivkovic
- Department of Pathology, Medical Faculty, University of Nis, Blvd. dr Zorana Djindjica 81, 18000 Nis, Serbia
| | - Ana Cvetanovic
- Clinic of Oncology, Clinical Center Nis, Blvd. dr Zorana Djindjica 48, 18000 Nis, Serbia
| | - Ivana Stojanovic
- Department of Biochemistry, Medical Faculty, University of Nis, Blvd. dr Zorana Djindjica 81, 18000 Nis, Serbia
| | - Miodrag Colic
- Department of Immunology, Medical Faculty, University of Nis, Blvd. dr Zorana Djindjica 81, 18000 Nis, Serbia
| |
Collapse
|
43
|
Chamberlain KA, Nanescu SE, Psachoulia K, Huang JK. Oligodendrocyte regeneration: Its significance in myelin replacement and neuroprotection in multiple sclerosis. Neuropharmacology 2016; 110:633-643. [PMID: 26474658 PMCID: PMC4841742 DOI: 10.1016/j.neuropharm.2015.10.010] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Revised: 09/22/2015] [Accepted: 10/05/2015] [Indexed: 12/12/2022]
Abstract
Oligodendrocytes readily regenerate and replace myelin membranes around axons in the adult mammalian central nervous system (CNS) following injury. The ability to regenerate oligodendrocytes depends on the availability of neural progenitors called oligodendrocyte precursor cells (OPCs) in the adult CNS that respond to injury-associated signals to induce OPC expansion followed by oligodendrocyte differentiation, axonal contact and myelin regeneration (remyelination). Remyelination ensures the maintenance of axonal conduction, and the oligodendrocytes themselves provide metabolic factors that are necessary to maintain neuronal integrity. Recent advances in oligodendrocyte regeneration research are beginning to shed light on critical intrinsic signals, as well as extrinsic, environmental factors that regulate the distinct steps of oligodendrocyte lineage progression and myelin replacement under CNS injury. These studies may offer novel pharmacological targets for regenerative medicine in inflammatory demyelinating disorders in the CNS such as multiple sclerosis. This article is part of the Special Issue entitled 'Oligodendrocytes in Health and Disease'.
Collapse
Affiliation(s)
- Kelly A Chamberlain
- Department of Biology, Georgetown University, Washington, D.C., USA; Interdisciplinary Program in Neuroscience, Georgetown University, Washington, D.C., USA
| | - Sonia E Nanescu
- Department of Biology, Georgetown University, Washington, D.C., USA
| | | | - Jeffrey K Huang
- Department of Biology, Georgetown University, Washington, D.C., USA; Interdisciplinary Program in Neuroscience, Georgetown University, Washington, D.C., USA.
| |
Collapse
|
44
|
Herwerth M, Kalluri SR, Srivastava R, Kleele T, Kenet S, Illes Z, Merkler D, Bennett JL, Misgeld T, Hemmer B. In vivo imaging reveals rapid astrocyte depletion and axon damage in a model of neuromyelitis optica-related pathology. Ann Neurol 2016; 79:794-805. [PMID: 26946517 PMCID: PMC5021140 DOI: 10.1002/ana.24630] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Revised: 03/02/2016] [Accepted: 03/03/2016] [Indexed: 01/27/2023]
Abstract
Objective Neuromyelitis optica (NMO) is an autoimmune disease of the central nervous system, which resembles multiple sclerosis (MS). NMO differs from MS, however, in the distribution and histology of neuroinflammatory lesions and shows a more aggressive clinical course. Moreover, the majority of NMO patients carry immunoglobulin G autoantibodies against aquaporin‐4 (AQP4), an astrocytic water channel. Antibodies against AQP4 can damage astrocytes by complement, but NMO histopathology also shows demyelination, and — importantly—axon injury, which may determine permanent deficits following NMO relapses. The dynamics of astrocyte injury in NMO and the mechanisms by which toxicity spreads to axons are not understood. Methods Here, we establish in vivo imaging of the spinal cord, one of the main sites of NMO pathology, as a powerful tool to study the formation of experimental NMO‐related lesions caused by human AQP4 antibodies in mice. Results We found that human AQP4 antibodies caused acute astrocyte depletion with initial oligodendrocyte survival. Within 2 hours of antibody application, we observed secondary axon injury in the form of progressive swellings. Astrocyte toxicity and axon damage were dependent on AQP4 antibody titer and complement, specifically C1q. Interpretation In vivo imaging of the spinal cord reveals the swift development of NMO‐related acute axon injury after AQP4 antibody‐mediated astrocyte depletion. This approach will be useful in studying the mechanisms underlying the spread of NMO pathology beyond astrocytes, as well as in evaluating potential neuroprotective interventions. Ann Neurol 2016;79:794–805
Collapse
Affiliation(s)
- Marina Herwerth
- Department of Neurology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.,Institute of Neuronal Cell Biology, Technical University of Munich, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Sudhakar Reddy Kalluri
- Department of Neurology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Rajneesh Srivastava
- Department of Neurology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Tatjana Kleele
- Institute of Neuronal Cell Biology, Technical University of Munich, Munich, Germany
| | - Selin Kenet
- Institute of Neuronal Cell Biology, Technical University of Munich, Munich, Germany
| | - Zsolt Illes
- Department of Neurology, Division of Clinical and Experimental Neuroimmunology, University of Pecs, Pecs, Hungary.,Department of Neurology and Institute of Clinical Research, Odense University Hospital, University of Southern Denmark, Odense, Denmark
| | - Doron Merkler
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland.,Division of Clinical Pathology, Geneva University Hospital, Geneva, Switzerland.,Department of Neuropathology, University Medical Center, Göttingen, Germany
| | - Jeffrey L Bennett
- Departments of Neurology and Ophthalmology, Program in Neuroscience, University of Colorado Denver School of Medicine, Aurora, CO
| | - Thomas Misgeld
- Institute of Neuronal Cell Biology, Technical University of Munich, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.,German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Center of Integrated Protein Science (CIPSM), Munich, Germany.,equal contributing senior authors
| | - Bernhard Hemmer
- Department of Neurology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.,equal contributing senior authors
| |
Collapse
|
45
|
Takagishi Y, Katanosaka K, Mizoguchi H, Murata Y. Disrupted axon-glia interactions at the paranode in myelinated nerves cause axonal degeneration and neuronal cell death in the aged Caspr mutant mouse shambling. Neurobiol Aging 2016; 43:34-46. [PMID: 27255813 DOI: 10.1016/j.neurobiolaging.2016.03.020] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 03/15/2016] [Accepted: 03/16/2016] [Indexed: 12/16/2022]
Abstract
Emerging evidence suggests that axonal degeneration is a disease mechanism in various neurodegenerative diseases and that the paranodes at the nodes of Ranvier may be the initial site of pathogenesis. We investigated the pathophysiology of the disease process in the central and peripheral nervous systems of a Caspr mutant mouse, shambling (shm), which is affected by disrupted paranodal structures and impaired nerve conduction of myelinated nerves. The shm mice manifest a progressive neurological phenotype as mice age. We found extensive axonal degeneration and a loss of neurons in the central nervous system and peripheral nervous system in aged shm mice. Axonal alteration of myelinated nerves was defined by abnormal distribution and expression of neurofilaments and derangements in the status of phosphorylated and non/de-phosphorylated neurofilaments. Autophagy-related structures were also accumulated in degenerated axons and neurons. In conclusion, our results suggest that disrupted axon-glia interactions at the paranode cause the cytoskeletal alteration in myelinated axons leading to neuronal cell death, and the process involves detrimental autophagy and aging as factors that promote the pathogenesis.
Collapse
Affiliation(s)
- Yoshiko Takagishi
- Department of Genetics, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan.
| | - Kimiaki Katanosaka
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
| | - Hiroyuki Mizoguchi
- Research Center for Next-Generation Drug Development, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
| | - Yoshiharu Murata
- Department of Genetics, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
| |
Collapse
|
46
|
Herold S, Kumar P, Wichert SP, Kretzschmar B, Bähr M, Rossner MJ, Hein K. Neurodegeneration in Autoimmune Optic Neuritis Is Associated with Altered APP Cleavage in Neurons and Up-Regulation of p53. PLoS One 2015; 10:e0138852. [PMID: 26426258 PMCID: PMC4591258 DOI: 10.1371/journal.pone.0138852] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 09/06/2015] [Indexed: 11/19/2022] Open
Abstract
Multiple Sclerosis (MS) is a chronic autoimmune inflammatory disease of the central nervous system (CNS). Histopathological and radiological analysis revealed that neurodegeneration occurs early in the disease course. However, the pathological mechanisms involved in neurodegeneration are poorly understood. Myelin oligodendrocyte glycoprotein (MOG)-induced experimental autoimmune encephalomyelitis (EAE) in Brown Norway rats (BN-rats) is a well-established animal model, especially of the neurodegenerative aspects of MS. Previous studies in this animal model indicated that loss of retinal ganglion cells (RGCs), the neurons that form the axons of the optic nerve, occurs in the preclinical phase of the disease and is in part independent of overt histopathological changes of the optic nerve. Therefore, the aim of this study was to identify genes which are involved in neuronal cell loss at different disease stages of EAE. Furthermore, genes that are highly specific for autoimmune-driven neurodegeneration were compared to those regulated in RGCs after optic nerve axotomy at corresponding time points. Using laser capture micro dissection we isolated RNA from unfixed RGCs and performed global transcriptome analysis of retinal neurons. In total, we detected 582 genes sequentially expressed in the preclinical phase and 1150 genes in the clinical manifest EAE (P < 0.05, fold-induction >1.5). Furthermore, using ingenuity pathway analysis (IPA), we identified amyloid precursor protein (APP) as a potential upstream regulator of changes in gene expression in the preclinical EAE but neither in clinical EAE, nor at any time point after optic nerve transection. Therefore, the gene pathway analysis lead to the hypothesis that altered cleavage of APP in neurons in the preclinical phase of EAE leads to the enhanced production of APP intracellular domain (AICD), which in turn acts as a transcriptional regulator and thereby initiates an apoptotic signaling cascade via up-regulation of the target gene p53.
Collapse
Affiliation(s)
- Sabine Herold
- Department of Neurology, University Medicine Göttingen, Göttingen, Germany
| | - Prateek Kumar
- Department of Neurology, University Medicine Göttingen, Göttingen, Germany
| | - Sven P. Wichert
- Molecular and Behavioral Neurobiology, Department of Psychiatry, Ludwig Maximilians University Munich, Munich, Germany
| | | | - Mathias Bähr
- Department of Neurology, University Medicine Göttingen, Göttingen, Germany
| | - Moritz J. Rossner
- Molecular and Behavioral Neurobiology, Department of Psychiatry, Ludwig Maximilians University Munich, Munich, Germany
| | - Katharina Hein
- Department of Neurology, University Medicine Göttingen, Göttingen, Germany
- * E-mail:
| |
Collapse
|
47
|
Yshii L, Gebauer C, Bernard-Valnet R, Liblau R. Neurons and T cells: Understanding this interaction for inflammatory neurological diseases. Eur J Immunol 2015; 45:2712-20. [DOI: 10.1002/eji.201545759] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 08/26/2015] [Accepted: 09/02/2015] [Indexed: 12/24/2022]
Affiliation(s)
- Lidia Yshii
- INSERM U1043 - CNRS UMR 5282; Centre de Physiopathologie Toulouse-Purpan; Toulouse France
- Université Toulouse III; Toulouse France
- Institute of Biomedical Sciences I; University of Sao Paulo; Sao Paulo Brazil
| | - Christina Gebauer
- INSERM U1043 - CNRS UMR 5282; Centre de Physiopathologie Toulouse-Purpan; Toulouse France
- Université Toulouse III; Toulouse France
| | - Raphaël Bernard-Valnet
- INSERM U1043 - CNRS UMR 5282; Centre de Physiopathologie Toulouse-Purpan; Toulouse France
- Université Toulouse III; Toulouse France
| | - Roland Liblau
- INSERM U1043 - CNRS UMR 5282; Centre de Physiopathologie Toulouse-Purpan; Toulouse France
- Université Toulouse III; Toulouse France
| |
Collapse
|
48
|
Roberts RA, Eitas TK, Byrne JD, Johnson BM, Short PJ, McKinnon KP, Reisdorf S, Luft JC, DeSimone JM, Ting JP. Towards programming immune tolerance through geometric manipulation of phosphatidylserine. Biomaterials 2015; 72:1-10. [PMID: 26325217 DOI: 10.1016/j.biomaterials.2015.08.040] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 08/17/2015] [Accepted: 08/18/2015] [Indexed: 12/22/2022]
Abstract
The possibility of engineering the immune system in a targeted fashion using biomaterials such as nanoparticles has made considerable headway in recent years. However, little is known as to how modulating the spatial presentation of a ligand augments downstream immune responses. In this report we show that geometric manipulation of phosphatidylserine (PS) through fabrication on rod-shaped PLGA nanoparticles robustly dampens inflammatory responses from innate immune cells while promoting T regulatory cell abundance by impeding effector T cell expansion. This response depends on the geometry of PS presentation as both PS liposomes and 1 micron cylindrical PS-PLGA particles are less potent signal inducers than 80 × 320 nm rod-shaped PS-PLGA particles for an equivalent dose of PS. We show that this immune tolerizing effect can be co-opted for therapeutic benefit in a mouse model of multiple sclerosis and an assay of organ rejection using a mixed lymphocyte reaction with primary human immune cells. These data provide evidence that geometric manipulation of a ligand via biomaterials may enable more efficient and tunable programming of cellular signaling networks for therapeutic benefit in a variety of disease states, including autoimmunity and organ rejection, and thus should be an active area of further research.
Collapse
Affiliation(s)
- Reid A Roberts
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Timothy K Eitas
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - James D Byrne
- Eshelman School of Pharmacy, Division of Molecular Pharmaceutics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Brandon M Johnson
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Patrick J Short
- Department of Chemistry, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Karen P McKinnon
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Shannon Reisdorf
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - J Christopher Luft
- Eshelman School of Pharmacy, Division of Molecular Pharmaceutics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Joseph M DeSimone
- Eshelman School of Pharmacy, Division of Molecular Pharmaceutics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Chemistry, University of North Carolina, Chapel Hill, NC 27599, USA
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC 27695, USA
- Sloan-Kettering Institute for Cancer Research, Memorial Sloan Kettering Comprehensive Cancer Center, New York, NY 10065, USA
| | - Jenny P Ting
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Center for Translational Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Institute for Inflammatory Diseases, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
49
|
Fressinaud C, Eyer J. Neurofilaments and NFL-TBS.40–63 peptide penetrate oligodendrocytes through clathrin-dependent endocytosis to promote their growth and survival in vitro. Neuroscience 2015; 298:42-51. [DOI: 10.1016/j.neuroscience.2015.04.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 03/05/2015] [Accepted: 04/02/2015] [Indexed: 12/13/2022]
|
50
|
Zhang W, Wang H, Zhang H, Leak RK, Shi Y, Hu X, Gao Y, Chen J. Dietary supplementation with omega-3 polyunsaturated fatty acids robustly promotes neurovascular restorative dynamics and improves neurological functions after stroke. Exp Neurol 2015; 272:170-80. [PMID: 25771800 DOI: 10.1016/j.expneurol.2015.03.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 03/02/2015] [Accepted: 03/06/2015] [Indexed: 12/12/2022]
Abstract
Stroke is a devastating neurological disease with no satisfactory therapies to preserve long-term neurological function, perhaps due to the sole emphasis on neuronal survival in most preclinical studies. Recent studies have revealed the importance of protecting multiple cell types in the injured brain, such as oligodendrocytes and components of the neurovascular unit, before long-lasting recovery of function can be achieved. For example, revascularization in the ischemic penumbra is critical to provide various neurotrophic factors that enhance the survival and activity of neurons and other progenitor cells, such as oligodendrocyte precursor cells. In the present study, we hypothesized that chronic dietary supplementation with fish oil promotes post-stroke angiogenesis, neurogenesis, and oligodendrogenesis, thereby leading to long-term functional improvements. Mice received dietary supplementation with n-3 PUFA-enriched fish oil for three months before and up to one month after stroke. As expected, dietary n-3 PUFAs significantly increased levels of n-3 PUFAs in the brain and improved long-term behavioral outcomes after cerebral ischemia. n-3 PUFAs also robustly improved revascularization and angiogenesis and boosted the survival of NeuN/BrdU labeled newborn neurons up to 35days after stroke injury. Furthermore, these pro-neurogenic effects were accompanied by robust oligodendrogenesis. Thus, this is the first study to demonstrate that chronic dietary intake of n-3 PUFAs is an effective prophylactic measure not only to protect against ischemic injury for the long term but also to actively promote neurovascular restorative dynamics and brain repair.
Collapse
Affiliation(s)
- Wenting Zhang
- State Key Laboratory of Medical Neurobiology and Institute of Brain Sciences, and Departments of Anesthesiology and Neurology of Huashan Hospital, Fudan University, Shanghai 200032, China; Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.
| | - Hailian Wang
- State Key Laboratory of Medical Neurobiology and Institute of Brain Sciences, and Departments of Anesthesiology and Neurology of Huashan Hospital, Fudan University, Shanghai 200032, China
| | - Hui Zhang
- State Key Laboratory of Medical Neurobiology and Institute of Brain Sciences, and Departments of Anesthesiology and Neurology of Huashan Hospital, Fudan University, Shanghai 200032, China
| | - Rehana K Leak
- Division of Pharmaceutical Sciences, Mylan School of Pharmacy, Duquesne University, Pittsburgh, PA 15282, USA
| | - Yejie Shi
- Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15261, USA
| | - Xiaoming Hu
- State Key Laboratory of Medical Neurobiology and Institute of Brain Sciences, and Departments of Anesthesiology and Neurology of Huashan Hospital, Fudan University, Shanghai 200032, China; Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15261, USA
| | - Yanqin Gao
- State Key Laboratory of Medical Neurobiology and Institute of Brain Sciences, and Departments of Anesthesiology and Neurology of Huashan Hospital, Fudan University, Shanghai 200032, China; Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Jun Chen
- State Key Laboratory of Medical Neurobiology and Institute of Brain Sciences, and Departments of Anesthesiology and Neurology of Huashan Hospital, Fudan University, Shanghai 200032, China; Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15261, USA.
| |
Collapse
|