1
|
Sekyi MT, Feri M, Desfor S, Atkinson KC, Golestany B, Beltran F, Tiwari-Woodruff SK. Demyelination and neurodegeneration early in experimental autoimmune encephalomyelitis contribute to functional deficits in the anterior visual pathway. Sci Rep 2024; 14:24048. [PMID: 39402114 PMCID: PMC11473523 DOI: 10.1038/s41598-024-73792-z] [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: 05/09/2024] [Accepted: 09/20/2024] [Indexed: 10/17/2024] Open
Abstract
Impaired visual function is a prevalent feature of optic neuritis (ON) in multiple sclerosis (MS). Abnormal visual evoked potential (VEP) findings of increased latencies, reduced amplitudes and abnormal waveforms as well as decreased retinal nerve fiber layer (RNFL) assessed by optical coherence tomography (OCT) are hallmarks of ON-induced visual dysfunction. Here we utilized the experimental autoimmune encephalomyelitis (EAE) mouse model of MS to investigate the functional and pathological progression during early (before any clinical symptoms), peak (initial maximal clinical symptoms), and late (chronic disease for > 3 weeks) disease stages. Demyelination and initial stages of axon damage were observed in early EAE. Significant demyelination, inflammation, increased axon damage and impaired P1/N2 amplitudes and latencies by VEP were seen in middle and late EAE groups. A decrease in RNFL thickness by OCT was observed only during late EAE. NanoString analysis of optic nerves from late EAE indicated elevated inflammation-related genes, reduced myelin-related genes, and changes in axon degeneration-related genes. Early inflammatory demyelination and functional deficits of the visual pathway, if untreated, may lead to severe irrecoverable axon damage in EAE. These studies potentially help explain the progression of visual dysfunction during MS.
Collapse
Affiliation(s)
- Maria T Sekyi
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Rm 3140, Multidisciplinary Research Building, 900 University Ave, Riverside, CA, 92521, USA
| | - Micah Feri
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Rm 3140, Multidisciplinary Research Building, 900 University Ave, Riverside, CA, 92521, USA
| | - Shane Desfor
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Rm 3140, Multidisciplinary Research Building, 900 University Ave, Riverside, CA, 92521, USA
| | - Kelley C Atkinson
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Rm 3140, Multidisciplinary Research Building, 900 University Ave, Riverside, CA, 92521, USA
| | - Batis Golestany
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Rm 3140, Multidisciplinary Research Building, 900 University Ave, Riverside, CA, 92521, USA
| | - Fernando Beltran
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Rm 3140, Multidisciplinary Research Building, 900 University Ave, Riverside, CA, 92521, USA
| | - Seema K Tiwari-Woodruff
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Rm 3140, Multidisciplinary Research Building, 900 University Ave, Riverside, CA, 92521, USA.
| |
Collapse
|
2
|
Olivero G, Taddeucci A, Vallarino G, Trebesova H, Roggeri A, Gagliani MC, Cortese K, Grilli M, Pittaluga A. Complement tunes glutamate release and supports synaptic impairments in an animal model of multiple sclerosis. Br J Pharmacol 2024. [PMID: 38369641 DOI: 10.1111/bph.16328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 12/05/2023] [Accepted: 01/03/2024] [Indexed: 02/20/2024] Open
Abstract
BACKGROUND AND PURPOSE To deepen our knowledge of the role of complement in synaptic impairment in experimental autoimmune encephalomyelitis (EAE) mice, we investigated the distribution of C1q and C3 proteins and the role of complement as a promoter of glutamate release in purified nerve endings (synaptosomes) and astrocytic processes (gliosomes) isolated from the cortex of EAE mice at the acute stage of the disease (21 ± 1 day post-immunization). EXPERIMENTAL APPROACH EAE cortical synaptosomes and gliosomes were analysed for glutamate release efficiency (measured as release of preloaded [3 H]D-aspartate ([3 H]D-ASP)), C1q and C3 protein density, and for viability and ongoing apoptosis. KEY RESULTS In healthy mice, complement releases [3 H]D-ASP from gliosomes more efficiently than from synaptosomes. The releasing activity occurs in a dilution-dependent manner and involves the reversal of the excitatory amino acid transporters (EAATs). In EAE mice, the complement-induced releasing activity is significantly reduced in cortical synaptosomes but amplified in cortical gliosomes. These adaptations are paralleled by decreased density of the EAAT2 protein in synaptosomes and increased EAAT1 staining in gliosomes. Concomitantly, PSD95, GFAP, and CD11b, but not SNAP25, proteins are overexpressed in the cortex of the EAE mice. Similarly, C1q and C3 protein immunostaining is increased in EAE cortical synaptosomes and gliosomes, although signs of ongoing apoptosis or altered viability are not detectable. CONCLUSION AND IMPLICATIONS Our results unveil a new noncanonical role of complement in the CNS of EAE mice relevant to disease progression and central synaptopathy that suggests new therapeutic targets for the management of MS.
Collapse
Affiliation(s)
- Guendalina Olivero
- Department of Pharmacy, DIFAR, Pharmacology and Toxicology Section, University of Genoa, Genoa, Italy
| | - Alice Taddeucci
- Department of Pharmacy, DIFAR, Pharmacology and Toxicology Section, University of Genoa, Genoa, Italy
| | - Giulia Vallarino
- Department of Pharmacy, DIFAR, Pharmacology and Toxicology Section, University of Genoa, Genoa, Italy
| | - Hanna Trebesova
- Department of Pharmacy, DIFAR, Pharmacology and Toxicology Section, University of Genoa, Genoa, Italy
| | - Alessandra Roggeri
- Department of Pharmacy, DIFAR, Pharmacology and Toxicology Section, University of Genoa, Genoa, Italy
| | - Maria Cristina Gagliani
- DIMES, Department of Experimental Medicine, Cellular Electron Microscopy Laboratory, Università di Genova, Genoa, Italy
| | - Katia Cortese
- DIMES, Department of Experimental Medicine, Cellular Electron Microscopy Laboratory, Università di Genova, Genoa, Italy
| | - Massimo Grilli
- Department of Pharmacy, DIFAR, Pharmacology and Toxicology Section, University of Genoa, Genoa, Italy
| | - Anna Pittaluga
- Department of Pharmacy, DIFAR, Pharmacology and Toxicology Section, Centre of Excellence for Biomedical Research, 3Rs Center, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| |
Collapse
|
3
|
Zhou R, Li J, Wang R, Chen Z, Zhou F. The neurovascular unit in healthy and injured spinal cord. J Cereb Blood Flow Metab 2023; 43:1437-1455. [PMID: 37190756 PMCID: PMC10414016 DOI: 10.1177/0271678x231172008] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 02/09/2023] [Accepted: 03/24/2023] [Indexed: 05/17/2023]
Abstract
The neurovascular unit (NVU) reflects the close temporal and spatial link between neurons and blood vessels. However, the understanding of the NVU in the spinal cord is far from clear and largely based on generalized knowledge obtained from the brain. Herein, we review the present knowledge of the NVU and highlight candidate approaches to investigate the NVU, particularly focusing on the spinal cord. Several unique features maintain the highly regulated microenvironment in the NVU. Autoregulation and neurovascular coupling ensure regional blood flow meets the metabolic demand according to the blood supply or local neural activation. The blood-central nervous system barrier partitions the circulating blood from neural parenchyma and facilitates the selective exchange of substances. Furthermore, we discuss spinal cord injury (SCI) as a common injury from the perspective of NVU dysfunction. Hopefully, this review will help expand the understanding of the NVU in the spinal cord and inspire new insights into SCI.
Collapse
Affiliation(s)
- Rubing Zhou
- Department of Orthopedics, Peking University Third Hospital, Beijing, China
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Junzhao Li
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Ruideng Wang
- Department of Orthopedics, Peking University Third Hospital, Beijing, China
| | - Zhengyang Chen
- Department of Orthopedics, Peking University Third Hospital, Beijing, China
| | - Fang Zhou
- Department of Orthopedics, Peking University Third Hospital, Beijing, China
| |
Collapse
|
4
|
Ma D, Sun C, Manne R, Guo T, Bosc C, Barry J, Magliery T, Andrieux A, Li H, Gu C. A cytoskeleton-membrane interaction conserved in fast-spiking neurons controls movement, emotion, and memory. Mol Psychiatry 2023; 28:3994-4010. [PMID: 37833406 PMCID: PMC10905646 DOI: 10.1038/s41380-023-02286-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 09/20/2023] [Accepted: 09/29/2023] [Indexed: 10/15/2023]
Abstract
The pathogenesis of schizophrenia is believed to involve combined dysfunctions of many proteins including microtubule-associated protein 6 (MAP6) and Kv3.1 voltage-gated K+ (Kv) channel, but their relationship and functions in behavioral regulation are often not known. Here we report that MAP6 stabilizes Kv3.1 channels in parvalbumin-positive (PV+ ) fast-spiking GABAergic interneurons, regulating behavior. MAP6-/- and Kv3.1-/- mice display similar hyperactivity and avoidance reduction. Their proteins colocalize in PV+ interneurons and MAP6 deletion markedly reduces Kv3.1 protein level. We further show that two microtubule-binding modules of MAP6 bind the Kv3.1 tetramerization domain with high affinity, maintaining the channel level in both neuronal soma and axons. MAP6 knockdown by AAV-shRNA in the amygdala or the hippocampus reduces avoidance or causes hyperactivity and recognition memory deficit, respectively, through elevating projection neuron activity. Finally, knocking down Kv3.1 or disrupting the MAP6-Kv3.1 binding in these brain regions causes avoidance reduction and hyperactivity, consistent with the effects of MAP6 knockdown. Thus, disrupting this conserved cytoskeleton-membrane interaction in fast-spiking neurons causes different degrees of functional vulnerability in various neural circuits.
Collapse
Affiliation(s)
- Di Ma
- Ohio State Biochemistry Program, The Ohio State University, Columbus, OH, USA
- Department of Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH, 43210, USA
| | - Chao Sun
- Department of Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH, 43210, USA
- MCDB graduate program, The Ohio State University, Columbus, OH, USA
| | - Rahul Manne
- Department of Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH, 43210, USA
| | - Tianqi Guo
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH, 43210, USA
| | - Christophe Bosc
- Univ. Grenoble Alpes, Inserm, U1216, CEA, Grenoble Institut Neurosciences, 38000, Grenoble, France
| | - Joshua Barry
- Department of Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH, 43210, USA
- IDDRC, Jane and Terry Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Thomas Magliery
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH, 43210, USA
| | - Annie Andrieux
- Univ. Grenoble Alpes, Inserm, U1216, CEA, Grenoble Institut Neurosciences, 38000, Grenoble, France
| | - Houzhi Li
- Department of Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH, 43210, USA
| | - Chen Gu
- Ohio State Biochemistry Program, The Ohio State University, Columbus, OH, USA.
- Department of Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH, 43210, USA.
- MCDB graduate program, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
5
|
Rashidbenam Z, Ozturk E, Pagnin M, Theotokis P, Grigoriadis N, Petratos S. How does Nogo receptor influence demyelination and remyelination in the context of multiple sclerosis? Front Cell Neurosci 2023; 17:1197492. [PMID: 37361998 PMCID: PMC10285164 DOI: 10.3389/fncel.2023.1197492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 05/15/2023] [Indexed: 06/28/2023] Open
Abstract
Multiple sclerosis (MS) can progress with neurodegeneration as a consequence of chronic inflammatory mechanisms that drive neural cell loss and/or neuroaxonal dystrophy in the central nervous system. Immune-mediated mechanisms can accumulate myelin debris in the disease extracellular milieu during chronic-active demyelination that can limit neurorepair/plasticity and experimental evidence suggests that potentiated removal of myelin debris can promote neurorepair in models of MS. The myelin-associated inhibitory factors (MAIFs) are integral contributors to neurodegenerative processes in models of trauma and experimental MS-like disease that can be targeted to promote neurorepair. This review highlights the molecular and cellular mechanisms that drive neurodegeneration as a consequence of chronic-active inflammation and outlines plausible therapeutic approaches to antagonize the MAIFs during the evolution of neuroinflammatory lesions. Moreover, investigative lines for translation of targeted therapies against these myelin inhibitors are defined with an emphasis on the chief MAIF, Nogo-A, that may demonstrate clinical efficacy of neurorepair during progressive MS.
Collapse
Affiliation(s)
- Zahra Rashidbenam
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Ezgi Ozturk
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Maurice Pagnin
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Paschalis Theotokis
- Laboratory of Experimental Neurology and Neuroimmunology, Department of Neurology, AHEPA University Hospital, Thessaloniki, Greece
| | - Nikolaos Grigoriadis
- Laboratory of Experimental Neurology and Neuroimmunology, Department of Neurology, AHEPA University Hospital, Thessaloniki, Greece
| | - Steven Petratos
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
6
|
Treatment with an antigen-specific dual microparticle system reverses advanced multiple sclerosis in mice. Proc Natl Acad Sci U S A 2022; 119:e2205417119. [PMID: 36256820 PMCID: PMC9618088 DOI: 10.1073/pnas.2205417119] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Antigen-specific therapies hold promise for treating autoimmune diseases such as multiple sclerosis while avoiding the deleterious side effects of systemic immune suppression due to delivering the disease-specific antigen as part of the treatment. In this study, an antigen-specific dual-sized microparticle (dMP) treatment reversed hind limb paralysis when administered in mice with advanced experimental autoimmune encephalomyelitis (EAE). Treatment reduced central nervous system (CNS) immune cell infiltration, demyelination, and inflammatory cytokine levels. Mechanistic insights using single-cell RNA sequencing showed that treatment impacted the MHC II antigen presentation pathway in dendritic cells, macrophages, B cells, and microglia, not only in the draining lymph nodes but also strikingly in the spinal cord. CD74 and cathepsin S were among the common genes down-regulated in most antigen presenting cell (APC) clusters, with B cells also having numerous MHC II genes reduced. Efficacy of the treatment diminished when B cells were absent, suggesting their impact in this therapy, in concert with other immune populations. Activation and inflammation were reduced in both APCs and T cells. This promising antigen-specific therapeutic approach advantageously engaged essential components of both innate and adaptive autoimmune responses and capably reversed paralysis in advanced EAE without the use of a broad immunosuppressant.
Collapse
|
7
|
The Mechanical Microenvironment Regulates Axon Diameters Visualized by Cryo-Electron Tomography. Cells 2022; 11:cells11162533. [PMID: 36010609 PMCID: PMC9406316 DOI: 10.3390/cells11162533] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/07/2022] [Accepted: 08/10/2022] [Indexed: 12/21/2022] Open
Abstract
Axonal varicosities or swellings are enlarged structures along axon shafts and profoundly affect action potential propagation and synaptic transmission. These structures, which are defined by morphology, are highly heterogeneous and often investigated concerning their roles in neuropathology, but why they are present in the normal brain remains unknown. Combining confocal microscopy and cryo-electron tomography (Cryo-ET) with in vivo and in vitro systems, we report that non-uniform mechanical interactions with the microenvironment can lead to 10-fold diameter differences within an axon of the central nervous system (CNS). In the brains of adult Thy1-YFP transgenic mice, individual axons in the cortex displayed significantly higher diameter variation than those in the corpus callosum. When being cultured on lacey carbon film-coated electron microscopy (EM) grids, CNS axons formed varicosities exclusively in holes and without microtubule (MT) breakage, and they contained mitochondria, multivesicular bodies (MVBs), and/or vesicles, similar to the axonal varicosities induced by mild fluid puffing. Moreover, enlarged axon branch points often contain MT free ends leading to the minor branch. When the axons were fasciculated by mimicking in vivo axonal bundles, their varicosity levels reduced. Taken together, our results have revealed the extrinsic regulation of the three-dimensional ultrastructures of central axons by the mechanical microenvironment under physiological conditions.
Collapse
|
8
|
Turner MP, Zhao Y, Abdelkarim D, Liu P, Spence JS, Hutchison JL, Sivakolundu DK, Thomas BP, Hubbard NA, Xu C, Taneja K, Lu H, Rypma B. Altered linear coupling between stimulus-evoked blood flow and oxygen metabolism in the aging human brain. Cereb Cortex 2022; 33:135-151. [PMID: 35388407 PMCID: PMC9758587 DOI: 10.1093/cercor/bhac057] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 01/13/2022] [Accepted: 01/14/2022] [Indexed: 11/14/2022] Open
Abstract
Neural-vascular coupling (NVC) is the process by which oxygen and nutrients are delivered to metabolically active neurons by blood vessels. Murine models of NVC disruption have revealed its critical role in healthy neural function. We hypothesized that, in humans, aging exerts detrimental effects upon the integrity of the neural-glial-vascular system that underlies NVC. To test this hypothesis, calibrated functional magnetic resonance imaging (cfMRI) was used to characterize age-related changes in cerebral blood flow (CBF) and oxygen metabolism during visual cortex stimulation. Thirty-three younger and 27 older participants underwent cfMRI scanning during both an attention-controlled visual stimulation task and a hypercapnia paradigm used to calibrate the blood-oxygen-level-dependent signal. Measurement of stimulus-evoked blood flow and oxygen metabolism permitted calculation of the NVC ratio to assess the integrity of neural-vascular communication. Consistent with our hypothesis, we observed monotonic NVC ratio increases with increasing visual stimulation frequency in younger adults but not in older adults. Age-related changes in stimulus-evoked cerebrovascular and neurometabolic signal could not fully explain this disruption; increases in stimulus-evoked neurometabolic activity elicited corresponding increases in stimulus-evoked CBF in younger but not in older adults. These results implicate age-related, demand-dependent failures of the neural-glial-vascular structures that comprise the NVC system.
Collapse
Affiliation(s)
- Monroe P Turner
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080, USA,Center for BrainHealth, University of Texas at Dallas, Dallas, TX, 75235, USA
| | - Yuguang Zhao
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080, USA,Center for BrainHealth, University of Texas at Dallas, Dallas, TX, 75235, USA
| | - Dema Abdelkarim
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080, USA,Center for BrainHealth, University of Texas at Dallas, Dallas, TX, 75235, USA
| | - Peiying Liu
- Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD 21287, USA
| | - Jeffrey S Spence
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080, USA,Center for BrainHealth, University of Texas at Dallas, Dallas, TX, 75235, USA
| | - Joanna L Hutchison
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080, USA,Center for BrainHealth, University of Texas at Dallas, Dallas, TX, 75235, USA
| | - Dinesh K Sivakolundu
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080, USA,Department of Biological Sciences, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Binu P Thomas
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA
| | - Nicholas A Hubbard
- Department of Psychology, Center for Brain, Biology, and Behavior, University of Nebraska, Lincoln, NE 68588, USA
| | - Cuimei Xu
- Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD 21287, USA
| | - Kamil Taneja
- Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD 21287, USA
| | - Hanzhang Lu
- Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD 21287, USA
| | - Bart Rypma
- Corresponding author: School of Behavioral and Brain Sciences, Center for Brain Health, University of Texas at Dallas, 800 West Campbell Road, Richardson, TX 75080, USA.
| |
Collapse
|
9
|
Sun C, Qi L, Cheng Y, Zhao Y, Gu C. Immediate induction of varicosities by transverse compression but not uniaxial stretch in axon mechanosensation. Acta Neuropathol Commun 2022; 10:7. [PMID: 35074017 PMCID: PMC8785443 DOI: 10.1186/s40478-022-01309-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 01/05/2022] [Indexed: 01/12/2023] Open
Abstract
Uniaxial stretch is believed to drive diffuse axonal injury (DAI) in mild traumatic brain injury (mTBI). Axonal varicosities are enlarged structures along axonal shafts and represent a hallmark feature of DAI. Here we report that axonal varicosities initiate in vivo immediately after head impact and are mainly induced by transverse compression but not uniaxial stretch. Vertical and lateral impacts to the mouse head induced axonal varicosities in distinct brain regions before any changes of microglial markers. Varicosities preferentially formed along axons perpendicular to impact direction. In cultured neurons, whereas 50% uniaxial strain was needed to rapidly induce axonal varicosities in a nanowrinkled stretch assay, physiologically-relevant transverse compression effectively induced axonal varicosities in a fluid puffing assay and can generate large but nonuniform deformation simulated by finite element analysis. Therefore, impact strength and direction may determine the threshold and spatial pattern of axonal varicosity initiation, respectively, partially resulting from intrinsic properties of axon mechanosensation.
Collapse
|
10
|
Schmaul S, Hanuscheck N, Bittner S. Astrocytic potassium and calcium channels as integrators of the inflammatory and ischemic CNS microenvironment. Biol Chem 2021; 402:1519-1530. [PMID: 34455729 DOI: 10.1515/hsz-2021-0256] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 08/13/2021] [Indexed: 12/24/2022]
Abstract
Astrocytes are key regulators of their surroundings by receiving and integrating stimuli from their local microenvironment, thereby regulating glial and neuronal homeostasis. Cumulating evidence supports a plethora of heterogenic astrocyte subpopulations that differ morphologically and in their expression patterns of receptors, transporters and ion channels, as well as in their functional specialisation. Astrocytic heterogeneity is especially relevant under pathological conditions. In experimental autoimmune encephalomyelitis (EAE), a mouse model of multiple sclerosis (MS), morphologically distinct astrocytic subtypes were identified and could be linked to transcriptome changes during different disease stages and regions. To allow for continuous awareness of changing stimuli across age and diseases, astrocytes are equipped with a variety of receptors and ion channels allowing the precise perception of environmental cues. Recent studies implicate the diverse repertoire of astrocytic ion channels - including transient receptor potential channels, voltage-gated calcium channels, inwardly rectifying K+ channels, and two-pore domain potassium channels - in sensing the brain state in physiology, inflammation and ischemia. Here, we review current evidence regarding astrocytic potassium and calcium channels and their functional contribution in homeostasis, neuroinflammation and stroke.
Collapse
Affiliation(s)
- Samantha Schmaul
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Centre of the Johannes Gutenberg University Mainz, Langenbeckstraße 1, D-55131 Mainz, Germany
| | - Nicholas Hanuscheck
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Centre of the Johannes Gutenberg University Mainz, Langenbeckstraße 1, D-55131 Mainz, Germany
| | - Stefan Bittner
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Centre of the Johannes Gutenberg University Mainz, Langenbeckstraße 1, D-55131 Mainz, Germany
| |
Collapse
|
11
|
Zhao Y, Liu P, Turner MP, Abdelkarim D, Lu H, Rypma B. The neural-vascular basis of age-related processing speed decline. Psychophysiology 2021; 58:e13845. [PMID: 34115388 DOI: 10.1111/psyp.13845] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 04/14/2021] [Accepted: 04/19/2021] [Indexed: 12/12/2022]
Abstract
Most studies examining neurocognitive aging are based on the blood-oxygen level-dependent signal obtained during functional magnetic resonance imaging (fMRI). The physiological basis of this signal is neural-vascular coupling, the process by which neurons signal cerebrovasculature to dilate in response to an increase in active neural metabolism due to stimulation. These fMRI studies of aging rely on the hemodynamic equivalence assumption that this process is not disrupted by physiologic deterioration associated with aging. Studies of neural-vascular coupling challenge this assumption and show that neural-vascular coupling is closely related to cognition. In this review, we put forward a theory of processing speed decline in aging and how it is related to age-related neural-vascular coupling changes based on the results of studies elucidating the relationships between cognition, cerebrovascular dynamics, and aging.
Collapse
Affiliation(s)
- Yuguang Zhao
- School of Behavioral and Brain Sciences, Center for Brain Health, University of Texas at Dallas, Richardson, TX, USA
| | - Peiying Liu
- School of Medicine, Department of Radiology, Johns Hopkins University, Baltimore, MD, USA
| | - Monroe P Turner
- School of Behavioral and Brain Sciences, Center for Brain Health, University of Texas at Dallas, Richardson, TX, USA
| | - Dema Abdelkarim
- School of Behavioral and Brain Sciences, Center for Brain Health, University of Texas at Dallas, Richardson, TX, USA
| | - Hanzhang Lu
- School of Medicine, Department of Radiology, Johns Hopkins University, Baltimore, MD, USA
| | - Bart Rypma
- School of Behavioral and Brain Sciences, Center for Brain Health, University of Texas at Dallas, Richardson, TX, USA
| |
Collapse
|
12
|
Bozic I, Savic D, Lavrnja I. Astrocyte phenotypes: Emphasis on potential markers in neuroinflammation. Histol Histopathol 2020; 36:267-290. [PMID: 33226087 DOI: 10.14670/hh-18-284] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Astrocytes, the most abundant glial cells in the central nervous system (CNS), have numerous integral roles in all CNS functions. They are essential for synaptic transmission and support neurons by providing metabolic substrates, secreting growth factors and regulating extracellular concentrations of ions and neurotransmitters. Astrocytes respond to CNS insults through reactive astrogliosis, in which they go through many functional and molecular changes. In neuroinflammatory conditions reactive astrocytes exert both beneficial and detrimental functions, depending on the context and heterogeneity of astrocytic populations. In this review we profile astrocytic diversity in the context of neuroinflammation; with a specific focus on multiple sclerosis (MS) and its best-described animal model experimental autoimmune encephalomyelitis (EAE). We characterize two main subtypes, protoplasmic and fibrous astrocytes and describe the role of intermediate filaments in the physiology and pathology of these cells. Additionally, we outline a variety of markers that are emerging as important in investigating astrocytic biology in both physiological conditions and neuroinflammation.
Collapse
Affiliation(s)
- Iva Bozic
- Institute for Biological Research "Sinisa Stankovic", National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Danijela Savic
- Institute for Biological Research "Sinisa Stankovic", National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Irena Lavrnja
- Institute for Biological Research "Sinisa Stankovic", National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia.
| |
Collapse
|
13
|
Lange Canhos L, Chen M, Falk S, Popper B, Straub T, Götz M, Sirko S. Repetitive injury and absence of monocytes promote astrocyte self-renewal and neurological recovery. Glia 2020; 69:165-181. [PMID: 32744730 DOI: 10.1002/glia.23893] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 07/07/2020] [Accepted: 07/08/2020] [Indexed: 12/24/2022]
Abstract
Unlike microglia and NG2 glia, astrocytes are incapable of migrating to sites of injury in the posttraumatic cerebral cortex, instead relying on proliferation to replenish their numbers and distribution in the affected region. However, neither the spectrum of their proliferative repertoire nor their postinjury distribution has been examined in vivo. Using a combination of different thymidine analogs and clonal analysis in a model of repetitive traumatic brain injury, we show for the first time that astrocytes that are quiescent following an initial injury can be coerced to proliferate after a repeated insult in the cerebral cortex grey matter. Interestingly, this process is promoted by invasion of monocytes to the injury site, as their genetic ablation (using CCR2-/- mice) increased the number of repetitively dividing astrocytes at the expense of newly proliferating astrocytes in repeatedly injured parenchyma. These differences profoundly affected both the distribution of astrocytes and recovery period for posttraumatic behavior deficits suggesting key roles of astrocyte self-renewal in brain repair after injury.
Collapse
Affiliation(s)
- Luisa Lange Canhos
- Physiological Genomics, Biomedical Center, Ludwig-Maximilians-University Munich, Munich, Germany.,Institute of Stem Cell Research, Helmholtz Zentrum Munich, Neuherberg, Germany.,Graduate School of Systemic Neurosciences (GSN-LMU), Ludwig-Maximilians-University Munich, Munich, Germany
| | - Muxin Chen
- Physiological Genomics, Biomedical Center, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Sven Falk
- Physiological Genomics, Biomedical Center, Ludwig-Maximilians-University Munich, Munich, Germany.,Institute of Stem Cell Research, Helmholtz Zentrum Munich, Neuherberg, Germany
| | - Bastian Popper
- Core Facility Animal Models, Biomedical Center, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Tobias Straub
- Core Facility Bioinformatics, Biomedical Center, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Magdalena Götz
- Physiological Genomics, Biomedical Center, Ludwig-Maximilians-University Munich, Munich, Germany.,Institute of Stem Cell Research, Helmholtz Zentrum Munich, Neuherberg, Germany.,Excellence Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Swetlana Sirko
- Physiological Genomics, Biomedical Center, Ludwig-Maximilians-University Munich, Munich, Germany.,Institute of Stem Cell Research, Helmholtz Zentrum Munich, Neuherberg, Germany
| |
Collapse
|
14
|
Sivakolundu DK, West KL, Zuppichini M, Turner MP, Abdelkarim D, Zhao Y, Spence JS, Lu H, Okuda DT, Rypma B. The neurovascular basis of processing speed differences in humans: A model-systems approach using multiple sclerosis. Neuroimage 2020; 215:116812. [PMID: 32276075 DOI: 10.1016/j.neuroimage.2020.116812] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 03/16/2020] [Accepted: 03/21/2020] [Indexed: 12/29/2022] Open
Abstract
Behavioral studies investigating fundamental cognitive abilities provide evidence that processing speed accounts for large proportions of performance variability between individuals. Processing speed decline is a hallmark feature of the cognitive disruption observed in healthy aging and in demyelinating diseases such as multiple sclerosis (MS), neuromyelitis optica, and Wilson's disease. Despite the wealth of evidence suggesting a central role for processing speed in cognitive decline, the neural mechanisms of this fundamental ability remain unknown. Intact neurovascular coupling, acute localized blood flow increases following neural activity, is essential for optimal neural function. We hypothesized that efficient coupling forms the neural basis of processing speed. Because MS features neural-glial-vascular system disruption, we used it as a model to test this hypothesis. To assess the integrity of the coupling system, we measured blood-oxygen-level-dependent (BOLD) signal in healthy controls (HCs) and MS patients using a 3T MRI scanner while they viewed radial checkerboards that flickered periodically at 8 Hz. To assess processing speed and cognitive function, we administered a battery of neuropsychological tests. While MS patients exhibited reduced ΔBOLD with reductions in processing speed, no such relationships were observed in HCs. To further investigate the mechanisms that underlie ΔBOLD-processing speed relationships, we assessed the physiologic components that constitute ΔBOLD signal (i.e., cerebral blood flow, ΔCBF; cerebral metabolic rate of oxygen, ΔCMRO2; neurovascular coupling ratio) in speed-preserved and -impaired MS patients. While ΔCBF and ΔCMRO2 showed no group-differences, the neurovascular coupling ratio was significantly reduced in speed-impaired MS patients compared to speed-preserved MS patients. Together, these results suggest that neurovascular uncoupling might underlie cognitive slowing in MS and might be the central pathogenic mechanism governing processing speed decline.
Collapse
Affiliation(s)
- Dinesh K Sivakolundu
- Center for BrainHealth, School of Behavioral and Brain Sciences, The University of Texas at Dallas, Dallas, TX, USA; Department of Biological Sciences, The University of Texas at Dallas, Dallas, TX, USA
| | - Kathryn L West
- Center for BrainHealth, School of Behavioral and Brain Sciences, The University of Texas at Dallas, Dallas, TX, USA
| | - Mark Zuppichini
- Center for BrainHealth, School of Behavioral and Brain Sciences, The University of Texas at Dallas, Dallas, TX, USA
| | - Monroe P Turner
- Center for BrainHealth, School of Behavioral and Brain Sciences, The University of Texas at Dallas, Dallas, TX, USA
| | - Dema Abdelkarim
- Center for BrainHealth, School of Behavioral and Brain Sciences, The University of Texas at Dallas, Dallas, TX, USA
| | - Yuguang Zhao
- Center for BrainHealth, School of Behavioral and Brain Sciences, The University of Texas at Dallas, Dallas, TX, USA
| | - Jeffrey S Spence
- Center for BrainHealth, School of Behavioral and Brain Sciences, The University of Texas at Dallas, Dallas, TX, USA
| | - Hanzhang Lu
- Department of Radiology, Johns Hopkins University, Baltimore, MD, USA
| | - Darin T Okuda
- Clinical Center for Multiple Sclerosis, Neuroinnovation Program, Multiple Sclerosis & Neuroimmunology Imaging Program, Department of Neurology & Neurotherapeutics, UT Southwestern Medical Center, Dallas, TX, USA
| | - Bart Rypma
- Center for BrainHealth, School of Behavioral and Brain Sciences, The University of Texas at Dallas, Dallas, TX, USA; Department of Psychiatry, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
15
|
Abdelkarim D, Zhao Y, Turner MP, Sivakolundu DK, Lu H, Rypma B. A neural-vascular complex of age-related changes in the human brain: Anatomy, physiology, and implications for neurocognitive aging. Neurosci Biobehav Rev 2019; 107:927-944. [DOI: 10.1016/j.neubiorev.2019.09.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 08/02/2019] [Accepted: 09/02/2019] [Indexed: 01/09/2023]
|
16
|
Rice J, Coutellier L, Weiner JL, Gu C. Region-specific interneuron demyelination and heightened anxiety-like behavior induced by adolescent binge alcohol treatment. Acta Neuropathol Commun 2019; 7:173. [PMID: 31703603 DOI: 10.1186/s40478-019-0829-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 10/13/2019] [Indexed: 12/12/2022] Open
Abstract
Adolescent binge drinking represents a major public health challenge and can lead to persistent neurological and mental conditions, but the underlying pathogenic mechanisms remain poorly understood. Using a mouse model of adolescent binge ethanol treatment (ABET), we found that this treatment induced behavioral changes associated with demyelination in different brain regions. After ABET, adolescent mice exhibited anxiogenic behaviors with no change in locomotion on the elevated plus maze, and impaired spatial memory indicated by a significant reduction in spontaneous alternation in the Y maze test. Both effects persisted into adulthood. Anatomical studies further showed that ABET induced a significant reduction of parvalbumin-positive (PV+) GABAergic interneurons and myelin density in the hippocampus and medial prefrontal cortex (mPFC). While these deficits in PV+ interneurons and myelin persisted into early adulthood in the hippocampus, the myelin density recovered in the mPFC. Moreover, whereas ABET mainly damaged myelin of PV+ axons in the hippocampus, it primarily damaged myelin of PV-negative axons in the mPFC. Thus, our findings reveal that an adolescent binge alcohol treatment regimen disrupts spatial working memory, increases anxiety-like behaviors, and exerts unique temporal and spatial patterns of gray matter demyelination in the hippocampus and mPFC.
Collapse
|
17
|
Synaptic alterations and immune response are sexually dimorphic in a non-pertussis toxin model of experimental autoimmune encephalomyelitis. Exp Neurol 2019; 323:113061. [PMID: 31499065 DOI: 10.1016/j.expneurol.2019.113061] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 08/16/2019] [Accepted: 09/05/2019] [Indexed: 12/30/2022]
Abstract
Multiple sclerosis is an autoimmune disorder of the central nervous system (CNS) characterized by locomotor impairments, cognitive deficits, affective disorders, and chronic pain. Females are predominately affected by MS compared to males and develop motor symptoms earlier. However, key symptoms affect all patients regardless of sex. Previous studies have shown that demyelination and axonal damage play key roles in symptom development, but it is unclear why sex differences exist in MS onset, and effective symptom treatment is still lacking. We here used a non-pertussis toxin (nPTX) experimental autoimmune encephalomyelitis (EAE) model in C57BL/6 mice, to explore chronic symptoms and sex differences in CNS autoimmunity. We observed that, like in humans, female mice developed motor disease earlier than males. Further, changes in pre- and post-synaptic protein expression levels were observed in a sexually dimorphic manner with an overall shift towards excitatory signaling. Our data suggest that this shift towards excitatory signaling is achieved through different mechanisms in males and females. Altogether, our study helps to better understand sex-specific disease mechanisms to ultimately develop better diagnostic and treatment tools.
Collapse
|
18
|
Nickel M, Eid F, Jukkola P, Gu C. Copper chelation and autoimmunity differentially impact myelin in the hippocampal-prefrontal circuit. J Neuroimmunol 2019; 334:576998. [PMID: 31254928 DOI: 10.1016/j.jneuroim.2019.576998] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 06/18/2019] [Accepted: 06/19/2019] [Indexed: 01/19/2023]
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system. About 50% of MS patients develop deficits in learning, memory and executive function, which are accompanied by demyelinating lesions in the hippocampus and/or prefrontal cortex (PFC). Why demyelination in these regions occurs in some patients but not in others and what is the underlying mechanism remain unclear. Here we report that myelin density in the hippocampus and PFC is markedly reduced in the cuprizone model, but not in the chronic experimental autoimmune encephalomyelitis. These two models can be used for studying different neuropathophysiological aspects of demyelinating diseases.
Collapse
Affiliation(s)
- Mara Nickel
- Department of Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH 43210, USA
| | - Farida Eid
- College of Arts and Sciences, The Ohio State University, Columbus, OH 43210, USA
| | - Peter Jukkola
- Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH 43210, USA
| | - Chen Gu
- Department of Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH 43210, USA; Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
19
|
Turner MP, Hubbard NA, Sivakolundu DK, Himes LM, Hutchison JL, Hart J, Spence JS, Frohman EM, Frohman TC, Okuda DT, Rypma B. Preserved canonicality of the BOLD hemodynamic response reflects healthy cognition: Insights into the healthy brain through the window of Multiple Sclerosis. Neuroimage 2019; 190:46-55. [PMID: 29454932 DOI: 10.1016/j.neuroimage.2017.12.081] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 12/18/2017] [Accepted: 12/22/2017] [Indexed: 10/18/2022] Open
Abstract
The hemodynamic response function (HRF), a model of brain blood-flow changes in response to neural activity, reflects communication between neurons and the vasculature that supplies these neurons in part by means of glial cell intermediaries (e.g., astrocytes). Intact neural-vascular communication might play a central role in optimal cognitive performance. This hypothesis can be tested by comparing healthy individuals to those with known white-matter damage and impaired performance, as seen in Multiple Sclerosis (MS). Glial cell intermediaries facilitate the ability of neurons to adequately convey metabolic needs to cerebral vasculature for sufficient oxygen and nutrient perfusion. In this study, we isolated measurements of the HRF that could quantify the extent to which white-matter affects neural-vascular coupling and cognitive performance. HRFs were modeled from multiple brain regions during multiple cognitive tasks using piecewise cubic spline functions, an approach that minimized assumptions regarding HRF shape that may not be valid for diseased populations, and were characterized using two shape metrics (peak amplitude and time-to-peak). Peak amplitude was reduced, and time-to-peak was longer, in MS patients relative to healthy controls. Faster time-to-peak was predicted by faster reaction time, suggesting an important role for vasodilatory speed in the physiology underlying processing speed. These results support the hypothesis that intact neural-glial-vascular communication underlies optimal neural and cognitive functioning.
Collapse
Affiliation(s)
- Monroe P Turner
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, USA
| | - Nicholas A Hubbard
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Dinesh K Sivakolundu
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, USA
| | - Lyndahl M Himes
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, USA
| | - Joanna L Hutchison
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, USA
| | - John Hart
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, USA; Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jeffrey S Spence
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, USA
| | - Elliot M Frohman
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Teresa C Frohman
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Darin T Okuda
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Bart Rypma
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, USA; Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
20
|
Cerebrovascular heterogeneity and neuronal excitability. Neurosci Lett 2018; 667:75-83. [DOI: 10.1016/j.neulet.2017.01.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2016] [Revised: 01/04/2017] [Accepted: 01/06/2017] [Indexed: 01/01/2023]
|
21
|
Jukkola P, Gu Y, Lovett-Racke AE, Gu C. Suppression of Inflammatory Demyelinaton and Axon Degeneration through Inhibiting Kv3 Channels. Front Mol Neurosci 2017; 10:344. [PMID: 29123469 PMCID: PMC5662905 DOI: 10.3389/fnmol.2017.00344] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 10/10/2017] [Indexed: 01/19/2023] Open
Abstract
The development of neuroprotective and repair strategies for treating progressive multiple sclerosis (MS) requires new insights into axonal injury. 4-aminopyridine (4-AP), a blocker of voltage-gated K+ (Kv) channels, is used in symptomatic treatment of progressive MS, but the underlying mechanism remains unclear. Here we report that deleting Kv3.1—the channel with the highest 4-AP sensitivity—reduces clinical signs in experimental autoimmune encephalomyelitis (EAE), a mouse model for MS. In Kv3.1 knockout (KO) mice, EAE lesions in sensory and motor tracts of spinal cord were markedly reduced, and radial astroglia were activated with increased expression of brain derived neurotrophic factor (BDNF). Kv3.3/Kv3.1 and activated BDNF receptors were upregulated in demyelinating axons in EAE and MS lesions. In spinal cord myelin coculture, BDNF treatment promoted myelination, and neuronal firing via altering channel expression. Therefore, suppressing Kv3.1 alters neural circuit activity, which may enhance BNDF signaling and hence protect axons from inflammatory insults.
Collapse
Affiliation(s)
- Peter Jukkola
- Biomedical Sciences Graduate Program, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| | - Yuanzheng Gu
- Department of Biological Chemistry and Pharmacology, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| | - Amy E Lovett-Racke
- Department of Microbial Infection and Immunity, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| | - Chen Gu
- Biomedical Sciences Graduate Program, Wexner Medical Center, The Ohio State University, Columbus, OH, United States.,Department of Biological Chemistry and Pharmacology, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
22
|
Gu Y, Jukkola P, Wang Q, Esparza T, Zhao Y, Brody D, Gu C. Polarity of varicosity initiation in central neuron mechanosensation. J Cell Biol 2017; 216:2179-2199. [PMID: 28606925 PMCID: PMC5496611 DOI: 10.1083/jcb.201606065] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 01/17/2017] [Accepted: 04/20/2017] [Indexed: 12/22/2022] Open
Abstract
Little is known about mechanical regulation of morphological and functional polarity of central neurons. In this study, we report that mechanical stress specifically induces varicosities in the axons but not the dendrites of central neurons by activating TRPV4, a Ca2+/Na+-permeable mechanosensitive channel. This process is unexpectedly rapid and reversible, consistent with the formation of axonal varicosities in vivo induced by mechanical impact in a mouse model of mild traumatic brain injury. In contrast, prolonged stimulation of glutamate receptors induces varicosities in dendrites but not in axons. We further show that axonal varicosities are induced by persistent Ca2+ increase, disassembled microtubules (MTs), and subsequently reversible disruption of axonal transport, and are regulated by stable tubulin-only polypeptide, an MT-associated protein. Finally, axonal varicosity initiation can trigger action potentials to antidromically propagate to the soma in retrograde signaling. Therefore, our study demonstrates a new feature of neuronal polarity: axons and dendrites preferentially respond to physical and chemical stresses, respectively.
Collapse
Affiliation(s)
- Yuanzheng Gu
- Department of Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH
| | - Peter Jukkola
- Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH
| | - Qian Wang
- Biomedical Engineering Department, The Ohio State University, Columbus, OH
| | - Thomas Esparza
- Department of Neurology, Washington University, St. Louis, MO
| | - Yi Zhao
- Biomedical Engineering Department, The Ohio State University, Columbus, OH
| | - David Brody
- Department of Neurology, Washington University, St. Louis, MO
| | - Chen Gu
- Department of Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH
- Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH
| |
Collapse
|
23
|
Affiliation(s)
- Wheaton T Little
- Takeda Pharmaceuticals, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan.
| | - Ceri H Davies
- Takeda Pharmaceuticals, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| |
Collapse
|
24
|
Occhieppo VB, Marchese NA, Rodríguez ID, Basmadjian OM, Baiardi G, Bregonzio C. Neurovascular unit alteration in somatosensory cortex and enhancement of thermal nociception induced by amphetamine involves central AT1receptor activation. Eur J Neurosci 2017; 45:1586-1593. [DOI: 10.1111/ejn.13594] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 04/20/2017] [Accepted: 04/21/2017] [Indexed: 12/20/2022]
Affiliation(s)
- Victoria Belén Occhieppo
- Instituto de Farmacología Experimental Córdoba (IFEC-CONICET) Departamento de Farmacología; Facultad de Ciencias Químicas Universidad Nacional de Córdoba; Edificio Nuevo de Ciencias I Ciudad Universitaria Córdoba; Haya de la Torre S/N, esquina Medina Allende Córdoba Argentina
| | - Natalia Andrea Marchese
- Instituto de Farmacología Experimental Córdoba (IFEC-CONICET) Departamento de Farmacología; Facultad de Ciencias Químicas Universidad Nacional de Córdoba; Edificio Nuevo de Ciencias I Ciudad Universitaria Córdoba; Haya de la Torre S/N, esquina Medina Allende Córdoba Argentina
| | - Iara Diamela Rodríguez
- Instituto de Farmacología Experimental Córdoba (IFEC-CONICET) Departamento de Farmacología; Facultad de Ciencias Químicas Universidad Nacional de Córdoba; Edificio Nuevo de Ciencias I Ciudad Universitaria Córdoba; Haya de la Torre S/N, esquina Medina Allende Córdoba Argentina
| | - Osvaldo Martin Basmadjian
- Instituto de Farmacología Experimental Córdoba (IFEC-CONICET) Departamento de Farmacología; Facultad de Ciencias Químicas Universidad Nacional de Córdoba; Edificio Nuevo de Ciencias I Ciudad Universitaria Córdoba; Haya de la Torre S/N, esquina Medina Allende Córdoba Argentina
| | - Gustavo Baiardi
- Laboratorio de Neurofarmacología (IIBYT-CONICET); Universidad Nacional de Córdoba Facultad de Ciencias Químicas; Universidad Católica de Córdoba; Córdoba Argentina
| | - Claudia Bregonzio
- Instituto de Farmacología Experimental Córdoba (IFEC-CONICET) Departamento de Farmacología; Facultad de Ciencias Químicas Universidad Nacional de Córdoba; Edificio Nuevo de Ciencias I Ciudad Universitaria Córdoba; Haya de la Torre S/N, esquina Medina Allende Córdoba Argentina
| |
Collapse
|
25
|
Park S, Guo Y, Jia X, Choe HK, Grena B, Kang J, Park J, Lu C, Canales A, Chen R, Yim YS, Choi GB, Fink Y, Anikeeva P. One-step optogenetics with multifunctional flexible polymer fibers. Nat Neurosci 2017; 20:612-619. [PMID: 28218915 PMCID: PMC5374019 DOI: 10.1038/nn.4510] [Citation(s) in RCA: 174] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 01/23/2017] [Indexed: 12/13/2022]
Abstract
Optogenetic interrogation of neural pathways relies on delivery of light-sensitive opsins into tissue and subsequent optical illumination and electrical recording from the regions of interest. Despite the recent development of multifunctional neural probes, integration of these modalities in a single biocompatible platform remains a challenge. We developed a device composed of an optical waveguide, six electrodes and two microfluidic channels produced via fiber drawing. Our probes facilitated injections of viral vectors carrying opsin genes while providing collocated neural recording and optical stimulation. The miniature (<200 μm) footprint and modest weight (<0.5 g) of these probes allowed for multiple implantations into the mouse brain, which enabled opto-electrophysiological investigation of projections from the basolateral amygdala to the medial prefrontal cortex and ventral hippocampus during behavioral experiments. Fabricated solely from polymers and polymer composites, these flexible probes minimized tissue response to achieve chronic multimodal interrogation of brain circuits with high fidelity.
Collapse
Affiliation(s)
- Seongjun Park
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Yuanyuan Guo
- Department of Material Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biomedical Engineering, Tohoku University, Sendai, Miyagi, Japan
- Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Xiaoting Jia
- Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Han Kyoung Choe
- McGovern Institute for Brain Research Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Benjamin Grena
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Material Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jeewoo Kang
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jiyeon Park
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Chi Lu
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Material Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Andres Canales
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Material Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ritchie Chen
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Material Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Yeong Shin Yim
- McGovern Institute for Brain Research Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Gloria B. Choi
- McGovern Institute for Brain Research Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Yoel Fink
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Material Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Polina Anikeeva
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Material Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
26
|
Oklinski MK, Skowronski MT, Skowronska A, Rützler M, Nørgaard K, Nieland JD, Kwon TH, Nielsen S. Aquaporins in the Spinal Cord. Int J Mol Sci 2016; 17:E2050. [PMID: 27941618 PMCID: PMC5187850 DOI: 10.3390/ijms17122050] [Citation(s) in RCA: 33] [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: 09/21/2016] [Revised: 11/16/2016] [Accepted: 11/25/2016] [Indexed: 12/11/2022] Open
Abstract
Aquaporins (AQPs) are water channel proteins robustly expressed in the central nervous system (CNS). A number of previous studies described the cellular expression sites and investigated their major roles and function in the brain and spinal cord. Among thirteen different mammalian AQPs, AQP1 and AQP4 have been mainly studied in the CNS and evidence has been presented that they play important roles in the pathogenesis of CNS injury, edema and multiple diseases such as multiple sclerosis, neuromyelitis optica spectrum disorders, amyotrophic lateral sclerosis, glioblastoma multiforme, Alzheimer's disease and Parkinson's disease. The objective of this review is to highlight the current knowledge about AQPs in the spinal cord and their proposed roles in pathophysiology and pathogenesis related to spinal cord lesions and injury.
Collapse
Affiliation(s)
- Michal K Oklinski
- Department of Health Science and Technology, Aalborg University, 9220 Aalborg, Denmark.
| | - Mariusz T Skowronski
- Department of Animal Physiology, University of Warmia and Mazury in Olsztyn, 10-752 Olsztyn, Poland.
| | - Agnieszka Skowronska
- Department of Human Physiology, University of Warmia and Mazury in Olsztyn, 10-752 Olsztyn, Poland.
| | - Michael Rützler
- Department of Health Science and Technology, Aalborg University, 9220 Aalborg, Denmark.
| | - Kirsten Nørgaard
- Department of Health Science and Technology, Aalborg University, 9220 Aalborg, Denmark.
| | - John D Nieland
- Department of Health Science and Technology, Aalborg University, 9220 Aalborg, Denmark.
| | - Tae-Hwan Kwon
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Taegu 41944, Korea.
| | - Søren Nielsen
- Department of Health Science and Technology, Aalborg University, 9220 Aalborg, Denmark.
| |
Collapse
|
27
|
Hubbard NA, Turner M, Hutchison JL, Ouyang A, Strain J, Oasay L, Sundaram S, Davis S, Remington G, Brigante R, Huang H, Hart J, Frohman T, Frohman E, Biswal BB, Rypma B. Multiple sclerosis-related white matter microstructural change alters the BOLD hemodynamic response. J Cereb Blood Flow Metab 2016; 36:1872-1884. [PMID: 26661225 PMCID: PMC5094308 DOI: 10.1177/0271678x15615133] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 09/15/2015] [Indexed: 01/21/2023]
Abstract
Multiple sclerosis (MS) results in inflammatory damage to white matter microstructure. Prior research using blood-oxygen-level dependent (BOLD) imaging indicates MS-related alterations to brain function. What is currently unknown is the extent to which white matter microstructural damage influences BOLD signal in MS. Here we assessed changes in parameters of the BOLD hemodynamic response function (HRF) in patients with relapsing-remitting MS compared to healthy controls. We also used diffusion tensor imaging to assess whether MS-related changes to the BOLD-HRF were affected by changes in white matter microstructural integrity. Our results showed MS-related reductions in BOLD-HRF peak amplitude. These MS-related amplitude decreases were influenced by individual differences in white matter microstructural integrity. Other MS-related factors including altered reaction time, limited spatial extent of BOLD activity, elevated lesion burden, or lesion proximity to regions of interest were not mediators of group differences in BOLD-HRF amplitude. Results are discussed in terms of functional hyperemic mechanisms and implications for analysis of BOLD signal differences.
Collapse
Affiliation(s)
- Nicholas A Hubbard
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, USA
| | - Monroe Turner
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, USA
| | - Joanna L Hutchison
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, USA.,Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Austin Ouyang
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jeremy Strain
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, USA
| | - Larry Oasay
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, USA
| | - Saranya Sundaram
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, USA
| | - Scott Davis
- Department of Applied Physiology and Wellness, Southern Methodist University, Dallas, TX, USA
| | - Gina Remington
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ryan Brigante
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, USA
| | - Hao Huang
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - John Hart
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, USA.,Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Teresa Frohman
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Elliot Frohman
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Bharat B Biswal
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - Bart Rypma
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, USA .,Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
28
|
Telmisartan-mediated metabolic profile conferred brain protection in diabetic hypertensive rats as evidenced by magnetic resonance imaging, behavioral studies and histology. Eur J Pharmacol 2016; 789:88-97. [PMID: 27417654 DOI: 10.1016/j.ejphar.2016.07.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 07/10/2016] [Accepted: 07/11/2016] [Indexed: 12/28/2022]
Abstract
Type 2 diabetes and hypertension are associated with cognitive dysfunction that includes pathological changes in brain tissue. It was speculated that the beneficial hypotensive effect of telmisartan, an angiotensin receptor 1 blocker, and its unique hypoglycemic effect due to its PPARγ-activation, could ameliorate the pathological changes in the brain that accompany these diseases. We examined the effect of telmisartan on brain changes in magnetic resonance imaging (MRI) T2-weighted scans, and behavioral and histological findings in the Cohen-Rosenthal Diabetic Hypertensive (CRDH) rat. Baseline and post-treatment values with telmisartan/vehicle (3 months) of blood pressure, blood glucose levels, behavioral tests, brain MRI scanning and immunohistological staining were obtained. Telmisartan significantly lowered blood pressure and blood glucose levels; induced consistent T2 reduction in specific gray and white regions including hippocampus, corpus callosum, amygdala and cortical regions; and significantly improved performance on behavioral tasks. Immunohistological analysis of the brain revealed significant amelioration of diabetes/hypertension-induced changes in white matter regions and microglia, evidenced by preserved myelin (LBF marker), and improved microglial neuronal markers GFAP, GAP43 and Iba1 expression. In conclusion, the behavioral performance, longitudinal MRI study and histology staining revealed the protective effects of telmisartan on brain microstructure and cognitive function.
Collapse
|
29
|
Lereim RR, Oveland E, Xiao Y, Torkildsen Ø, Wergeland S, Myhr KM, Sun SC, Berven FS. The Brain Proteome of the Ubiquitin Ligase Peli1 Knock-Out Mouse during Experimental Autoimmune Encephalomyelitis. ACTA ACUST UNITED AC 2016; 9:209-219. [PMID: 27746629 PMCID: PMC5061044 DOI: 10.4172/jpb.1000408] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The ubiquitin ligase Peli1 has previously been suggested as a potential treatment target in multiple sclerosis. In the multiple sclerosis disease model, experimental autoimmune encephalomyelitis, Peli1 knock-out led to less activated microglia and less inflammation in the central nervous system. Despite being important in microglia, Peli1 expression has also been detected in glial and neuronal cells. In the present study the overall brain proteomes of Peli1 knock-out mice and wild-type mice were compared prior to experimental autoimmune encephalomyelitis induction, at onset of the disease and at disease peak. Brain samples from the frontal hemisphere, peripheral from the extensive inflammatory foci, were analyzed using TMT-labeling of sample pools, and the discovered proteins were verified in individual mice using label-free proteomics. The greatest proteomic differences between Peli1 knock-out and wild-type mice were observed at the disease peak. In Peli1 knock-out a higher degree of antigen presentation, increased activity of adaptive and innate immune cells and alterations to proteins involved in iron metabolism were observed during experimental autoimmune encephalomyelitis. These results unravel global effects to the brain proteome when abrogating Peli1 expression, underlining the importance of Peli1 as a regulator of the immune response also peripheral to inflammatory foci during experimental autoimmune encephalomyelitis. The proteomics data is available in PRIDE with accession PXD003710.
Collapse
Affiliation(s)
- Ragnhild Reehorst Lereim
- Proteomics Unit, Department of Biomedicine, University of Bergen, Norway; Kristian Gerhard Jebsen MS Research Centre, Department of Clinical Medicine, University of Bergen, Bergen Norway
| | - Eystein Oveland
- Proteomics Unit, Department of Biomedicine, University of Bergen, Norway; Kristian Gerhard Jebsen MS Research Centre, Department of Clinical Medicine, University of Bergen, Bergen Norway
| | - Yichuan Xiao
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA; Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai Jiao Tong University, Shanghai 200031, China
| | - Øivind Torkildsen
- Kristian Gerhard Jebsen MS Research Centre, Department of Clinical Medicine, University of Bergen, Bergen Norway; Norwegian Multiple Sclerosis Competence Centre, Department of Neurology, Haukeland University Hospital, Bergen, Norway
| | - Stig Wergeland
- Kristian Gerhard Jebsen MS Research Centre, Department of Clinical Medicine, University of Bergen, Bergen Norway; Norwegian Multiple Sclerosis Competence Centre, Department of Neurology, Haukeland University Hospital, Bergen, Norway
| | - Kjell-Morten Myhr
- Kristian Gerhard Jebsen MS Research Centre, Department of Clinical Medicine, University of Bergen, Bergen Norway; Norwegian Multiple Sclerosis Competence Centre, Department of Neurology, Haukeland University Hospital, Bergen, Norway
| | - Shao-Cong Sun
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Frode S Berven
- Proteomics Unit, Department of Biomedicine, University of Bergen, Norway; Kristian Gerhard Jebsen MS Research Centre, Department of Clinical Medicine, University of Bergen, Bergen Norway; Norwegian Multiple Sclerosis Competence Centre, Department of Neurology, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
30
|
Abstract
Astrocytes are the most explored non-neuronal cells in the brain under neurophysiological and neurodegenerative conditions. Extensive research has been done to understand their specific role during neuropathological conditions but still the existing findings could not conclude their mechanism of action and their specific role in neurodegenerative conditions. This review discusses their physiological and pathological roles, their activation, morphological alterations and their probable use in search of new therapeutic targets for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Sarika Singh
- a 1 Toxicology Division, CSIR-CDRI , Lucknow , India.,b 2 Department of Biochemistry and Biophysics , University of California , San Francisco, San Francisco , CA , USA
| | - Neeraj Joshi
- a 1 Toxicology Division, CSIR-CDRI , Lucknow , India.,b 2 Department of Biochemistry and Biophysics , University of California , San Francisco, San Francisco , CA , USA
| |
Collapse
|
31
|
Jha MK, Lee WH, Suk K. Functional polarization of neuroglia: Implications in neuroinflammation and neurological disorders. Biochem Pharmacol 2015; 103:1-16. [PMID: 26556658 DOI: 10.1016/j.bcp.2015.11.003] [Citation(s) in RCA: 191] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2015] [Accepted: 11/02/2015] [Indexed: 12/15/2022]
Abstract
Recent neuroscience research has established the adult brain as a dynamic organ having a unique ability to undergo changes with time. Neuroglia, especially microglia and astrocytes, provide dynamicity to the brain. Activation of these glial cells is a major component of the neuroinflammatory responses underlying brain injury and neurodegeneration. Glial cells execute functional reaction programs in response to diverse microenvironmental signals manifested by neuropathological conditions. Activated microglia exist along a continuum of two functional states of polarization namely M1-type (classical/proinflammatory activation) and M2-type (alternative/anti-inflammatory activation) as in macrophages. The balance between classically and alternatively activated microglial phenotypes influences disease progression in the CNS. The classically activated state of microglia drives the neuroinflammatory response and mediates the detrimental effects on neurons, whereas in their alternative activation state, which is apparently a beneficial activation state, the microglia play a crucial role in tissue maintenance and repair. Likewise, in response to immune or inflammatory microenvironments astrocytes also adopt neurotoxic or neuroprotective phenotypes. Reactive astrocytes exhibit two distinctive functional phenotypes defined by pro- or anti-inflammatory gene expression profile. In this review, we have thoroughly covered recent advances in the understanding of the functional polarization of brain and peripheral glia and its implications in neuroinflammation and neurological disorders. The identifiable phenotypes adopted by neuroglia in response to specific insult or injury can be exploited as promising diagnostic markers of neuroinflammatory diseases. Furthermore, harnessing the beneficial effects of the polarized glia could undoubtedly pave the way for the formulation of novel glia-based therapeutic strategies for diverse neurological disorders.
Collapse
Affiliation(s)
- Mithilesh Kumar Jha
- Department of Pharmacology, Brain Science & Engineering Institute, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Won-Ha Lee
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea
| | - Kyoungho Suk
- Department of Pharmacology, Brain Science & Engineering Institute, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University School of Medicine, Daegu, Republic of Korea.
| |
Collapse
|
32
|
Jukkola P, Gu C. Regulation of neurovascular coupling in autoimmunity to water and ion channels. Autoimmun Rev 2015; 14:258-67. [PMID: 25462580 PMCID: PMC4303502 DOI: 10.1016/j.autrev.2014.11.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 11/15/2014] [Indexed: 12/27/2022]
Abstract
Much progress has been made in understanding autoimmune channelopathies, but the underlying pathogenic mechanisms are not always clear due to broad expression of some channel proteins. Recent studies show that autoimmune conditions that interfere with neurovascular coupling in the central nervous system (CNS) can lead to neurodegeneration. Cerebral blood flow that meets neuronal activity and metabolic demand is tightly regulated by local neural activity. This process of reciprocal regulation involves coordinated actions of a number of cell types, including neurons, glia, and vascular cells. In particular, astrocytic endfeet cover more than 90% of brain capillaries to assist blood-brain barrier (BBB) function, and wrap around synapses and nodes of Ranvier to communicate with neuronal activity. In this review, we highlight four types of channel proteins that are expressed in astrocytes, regarding their structures, biophysical properties, expression and distribution patterns, and related diseases including autoimmune disorders. Water channel aquaporin 4 (AQP4) and inwardly rectifying potassium (Kir4.1) channels are concentrated in astrocytic endfeet, whereas some voltage-gated Ca(2+) and two-pore domain K(+) channels are expressed throughout the cell body of reactive astrocytes. More channel proteins are found in astrocytes under normal and abnormal conditions. This research field will contribute to a better understanding of pathogenic mechanisms underlying autoimmune disorders.
Collapse
Affiliation(s)
- Peter Jukkola
- Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH 43210, USA
| | - Chen Gu
- Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH 43210, USA; Department of Neuroscience, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
33
|
Kandratavicius L, Peixoto-Santos JE, Monteiro MR, Scandiuzzi RC, Carlotti CG, Assirati JA, Hallak JE, Leite JP. Mesial temporal lobe epilepsy with psychiatric comorbidities: a place for differential neuroinflammatory interplay. J Neuroinflammation 2015; 12:38. [PMID: 25889039 PMCID: PMC4347571 DOI: 10.1186/s12974-015-0266-z] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2014] [Accepted: 02/10/2015] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Despite the strong association between epilepsy and psychiatric comorbidities, few biological substrates are currently described. We have previously reported neuropathological alterations in mesial temporal lobe epilepsy (MTLE) patients with major depression and psychosis that suggest a morphological and neurochemical basis for psychopathological symptoms. Neuroinflammatory-related structures and molecules might be part of the altered neurochemical milieu underlying the association between epilepsy and psychiatric comorbidities, and such features have not been previously investigated in humans. METHODS MTLE hippocampi of subjects without psychiatric history (MTLEW), MTLE + major depression (MTLE + D), and MTLE + interictal psychosis (MTLE + P) derived from epilepsy surgery and control necropsies were investigated for reactive astrocytes (glial fibrillary acidic protein (GFAP)), activated microglia (human leukocyte antigen, MHC class II (HLA-DR)), glial metallothionein-I/II (MT-I/II), and aquaporin 4 (AQP4) immunohistochemistry. RESULTS We found an increased GFAP immunoreactive area in the molecular layers, granule cell layer, and cornus ammonis region 2 (CA2) and cornus ammonis region 1 (CA1) of MTLEW and MTLE + P, respectively, compared to MTLE + D. HLA-DR immunoreactive area was higher in cornus ammonis region 3 (CA3) of MTLE + P, compared to MTLE + D and MTLEW, and in the hilus, when compared to MTLEW. MTLEW cases showed increased MT-I/II area in the granule cell layer and CA1, compared to MTLE + P, and in the parasubiculum, when compared to MTLE + D and MTLE + P. Differences between MTLE and control, such as astrogliosis, microgliosis, increased MT-I/II, and decreased perivascular AQP4 in the epileptogenic hippocampus, were in agreement to what is currently described in the literature. CONCLUSIONS Neuroinflammatory-related molecules in MTLE hippocampus show a distinct pattern of expression when patients present with a comorbid psychiatric diagnosis, similar to what is found in the pure forms of schizophrenia and major depression. Future studies focusing on inflammatory characteristics of MTLE with psychiatric comorbidities might help in the design of better therapeutic strategies.
Collapse
Affiliation(s)
- Ludmyla Kandratavicius
- Department of Neurosciences and Behavior, Ribeirao Preto Medical School, University of Sao Paulo (USP), Av Bandeirantes 3900, CEP 14049-900, Ribeirao Preto, SP, Brazil. .,Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), USP, Ribeirao Preto, Brazil.
| | - Jose Eduardo Peixoto-Santos
- Department of Neurosciences and Behavior, Ribeirao Preto Medical School, University of Sao Paulo (USP), Av Bandeirantes 3900, CEP 14049-900, Ribeirao Preto, SP, Brazil.
| | - Mariana Raquel Monteiro
- Department of Neurosciences and Behavior, Ribeirao Preto Medical School, University of Sao Paulo (USP), Av Bandeirantes 3900, CEP 14049-900, Ribeirao Preto, SP, Brazil.
| | - Renata Caldo Scandiuzzi
- Department of Neurosciences and Behavior, Ribeirao Preto Medical School, University of Sao Paulo (USP), Av Bandeirantes 3900, CEP 14049-900, Ribeirao Preto, SP, Brazil.
| | | | | | - Jaime Eduardo Hallak
- Department of Neurosciences and Behavior, Ribeirao Preto Medical School, University of Sao Paulo (USP), Av Bandeirantes 3900, CEP 14049-900, Ribeirao Preto, SP, Brazil. .,Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), USP, Ribeirao Preto, Brazil. .,National Institute of Science and Technology in Translational Medicine (INCT-TM - CNPq), Ribeirao Preto, Brazil.
| | - Joao Pereira Leite
- Department of Neurosciences and Behavior, Ribeirao Preto Medical School, University of Sao Paulo (USP), Av Bandeirantes 3900, CEP 14049-900, Ribeirao Preto, SP, Brazil. .,Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), USP, Ribeirao Preto, Brazil.
| |
Collapse
|
34
|
Prakash J, Chouhan S, Yadav SK, Westfall S, Rai SN, Singh SP. Withania somnifera alleviates parkinsonian phenotypes by inhibiting apoptotic pathways in dopaminergic neurons. Neurochem Res 2014; 39:2527-36. [PMID: 25403619 DOI: 10.1007/s11064-014-1443-7] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Revised: 09/22/2014] [Accepted: 09/23/2014] [Indexed: 10/24/2022]
Abstract
Maneb (MB) and paraquat (PQ) are environmental toxins that have been experimentally used to induce selective damage of dopaminergic neurons leading to the development of Parkinson's disease (PD). Although the mechanism of this selective neuronal toxicity in not fully understood, oxidative stress has been linked to the pathogenesis of PD. The present study investigates the mechanisms of neuroprotection elicited by Withania somnifera (Ws), a herb traditionally recognized by the Indian system of medicine, Ayurveda. An ethanolic root extract of Ws was co-treated with the MB-PQ induced mouse model of PD and was shown to significantly rescue canonical indicators of PD including compromised locomotor activity, reduced dopamine in the substantia nigra and various aspects of oxidative damage. In particular, Ws reduced the expression of iNOS, a measure of oxidative stress. Ws also significantly improved the MB + PQ mediated induction of a pro-apoptotic state by reducing Bax and inducing Bcl-2 protein expression, respectively. Finally, Ws reduced expression of the pro-inflammatory marker of astrocyte activation, GFAP. Altogether, the present study suggests that Ws treatment provides nigrostriatal dopaminergic neuroprotection against MB-PQ induced Parkinsonism by the modulation of oxidative stress and apoptotic machinery possibly accounting for the behavioural effects.
Collapse
Affiliation(s)
- Jay Prakash
- Department of Biochemistry, Faculty of Science, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India,
| | | | | | | | | | | |
Collapse
|
35
|
Barry J, Gu Y, Jukkola P, O'Neill B, Gu H, Mohler PJ, Rajamani KT, Gu C. Ankyrin-G directly binds to kinesin-1 to transport voltage-gated Na+ channels into axons. Dev Cell 2014; 28:117-31. [PMID: 24412576 DOI: 10.1016/j.devcel.2013.11.023] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Revised: 10/14/2013] [Accepted: 11/26/2013] [Indexed: 12/26/2022]
Abstract
Action potentials (APs) propagating along axons require the activation of voltage-gated Na(+) (Nav) channels. How Nav channels are transported into axons is unknown. We show that KIF5/kinesin-1 directly binds to ankyrin-G (AnkG) to transport Nav channels into axons. KIF5 and Nav1.2 channels bind to multiple sites in the AnkG N-terminal domain that contains 24 ankyrin repeats. Disrupting AnkG-KIF5 binding with small interfering RNA or dominant-negative constructs markedly reduced Nav channel levels at the axon initial segment (AIS) and along entire axons, thereby decreasing AP firing. Live-cell imaging showed that fluorescently tagged AnkG or Nav1.2 cotransported with KIF5 along axons. Deleting AnkG in vivo or virus-mediated expression of a dominant-negative KIF5 construct specifically decreased the axonal level of Nav, but not Kv1.2, channels in mouse cerebellum. These results indicate that AnkG functions as an adaptor to link Nav channels to KIF5 during axonal transport before anchoring them to the AIS and nodes of Ranvier.
Collapse
Affiliation(s)
- Joshua Barry
- Molecular, Cellular and Developmental Biology Graduate Program, The Ohio State University, Columbus, OH 43210, USA
| | - Yuanzheng Gu
- Department of Neuroscience, The Ohio State University, Columbus, OH 43210, USA
| | - Peter Jukkola
- Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH 43210, USA
| | - Brian O'Neill
- Department of Pharmacology, The Ohio State University, Columbus, OH 43210, USA
| | - Howard Gu
- Department of Pharmacology, The Ohio State University, Columbus, OH 43210, USA
| | - Peter J Mohler
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA; Departments of Internal Medicine and Physiology and Cell Biology, The Ohio State University, Columbus, OH 43210, USA
| | | | - Chen Gu
- Molecular, Cellular and Developmental Biology Graduate Program, The Ohio State University, Columbus, OH 43210, USA; Department of Neuroscience, The Ohio State University, Columbus, OH 43210, USA; Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|