1
|
Mireles-Ramírez MA, Pacheco-Moises FP, González-Usigli HA, Sánchez-Rosales NA, Hernández-Preciado MR, Delgado-Lara DLC, Hernández-Cruz JJ, Ortiz GG. Neuromyelitis optica spectrum disorder: pathophysiological approach. Int J Neurosci 2024; 134:826-838. [PMID: 36453541 DOI: 10.1080/00207454.2022.2153046] [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: 03/12/2022] [Revised: 11/19/2022] [Accepted: 11/24/2022] [Indexed: 12/02/2022]
Abstract
Aim: To review the main pathological findings of Neuromyelitis Optica Spectrum Disorder (NMOSD) associated with the presence of autoantibodies to aquaporin-4 (AQP4) as well as the mechanisms of astrocyte dysfunction and demyelination. Methods: An comprehensive search of the literature in the field was carried out using the database of The National Center for Biotechnology Information from . Systematic searches were performed until July 2022. Results: NMOSD is an inflammatory and demyelinating disease of the central nervous system mainly in the areas of the optic nerves and spinal cord, thus explaining mostly the clinical findings. Other areas affected in NMOSD are the brainstem, hypothalamus, and periventricular regions. Relapses in NMOSD are generally severe and patients only partially recover. NMOSD includes clinical conditions where autoantibodies to aquaporin-4 (AQP4-IgG) of astrocytes are detected as well as similar clinical conditions where such antibodies are not detected. AQP4 are channel-forming integral membrane proteins of which AQ4 isoforms are able to aggregate in supramolecular assemblies termed orthogonal arrays of particles (OAP) and are essential in the regulation of water homeostasis and the adequate modulation of neuronal activity and circuitry. AQP4 assembly in orthogonal arrays of particles is essential for AQP4-IgG pathogenicity since AQP4 autoantibodies bind to OAPs with higher affinity than for AQP4 tetramers. NMOSD has a complex background with prominent roles for genes encoding cytokines and cytokine receptors. AQP4 autoantibodies activate the complement-mediated inflammatory demyelination and the ensuing damage to AQP4 water channels, leading to water influx, necrosis and axonal loss. Conclusions: NMOSD as an astrocytopathy is a nosological entity different from multiple sclerosis with its own serological marker: immunoglobulin G-type autoantibodies against the AQP4 protein which elicits a complement-dependent cytotoxicity and neuroinflammation. Some patients with typical manifestations of NMSOD are AQP4 seronegative and myelin oligodendrocyte glycoprotein positive. Thus, the detection of autoantibodies against AQP4 or other autoantibodies is crucial for the correct treatment of the disease and immunosuppressant therapy is the first choice.
Collapse
Affiliation(s)
- Mario A Mireles-Ramírez
- Department of Neurology, High Specialty Medical Unit, Western National Medical Center of the Mexican Institute of Social Security, Guadalajara, Jalisco, Mexico
| | - Fermín P Pacheco-Moises
- Department of Chemistry, University Center of Exact Sciences and Engineering; University of Guadalajara, Guadalajara, Jalisco, Mexico
| | - Héctor A González-Usigli
- Department of Neurology, High Specialty Medical Unit, Western National Medical Center of the Mexican Institute of Social Security, Guadalajara, Jalisco, Mexico
| | - Nayeli A Sánchez-Rosales
- Department of Neurology, High Specialty Medical Unit, Western National Medical Center of the Mexican Institute of Social Security, Guadalajara, Jalisco, Mexico
| | - Martha R Hernández-Preciado
- Department of Neurology, High Specialty Medical Unit, Western National Medical Center of the Mexican Institute of Social Security, Guadalajara, Jalisco, Mexico
| | | | - José J Hernández-Cruz
- Department of Philosophical and Methodological Disciplines and Service of Molecular Biology in medicine HC, University Health Sciences Center, University of Guadalajara, Guadalajara, Jalisco, Mexico
| | - Genaro Gabriel Ortiz
- Department of Neurology, High Specialty Medical Unit, Western National Medical Center of the Mexican Institute of Social Security, Guadalajara, Jalisco, Mexico
- Department of Philosophical and Methodological Disciplines and Service of Molecular Biology in medicine HC, University Health Sciences Center, University of Guadalajara, Guadalajara, Jalisco, Mexico
| |
Collapse
|
2
|
Damasio A, Damasio H. Homeostatic Feelings and the Emergence of Consciousness. J Cogn Neurosci 2024; 36:1653-1659. [PMID: 38319678 DOI: 10.1162/jocn_a_02119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2024]
Abstract
In this article, we summarize our views on the problem of consciousness and outline the current version of a novel hypothesis for how conscious minds can be generated in mammalian organisms. We propose that a mind can be considered conscious when three processes are in place: the first is a continuous generation of interoceptive feelings, which results in experiencing of the organism's internal operations; the second is the equally continuous production of images, generated according to the organism's sensory perspective relative to its surround; the third combines feeling/experience and perspective resulting in a process of subjectivity relative to the image contents. We also propose a biological basis for these three components: the peripheral and central physiology of interoception and exteroception help explain the implementation of the first two components, whereas the third depends on central nervous system integration, at multiple levels, from spinal cord, brainstem, and diencephalic nuclei, to selected regions of the mesial cerebral cortices.
Collapse
|
3
|
Guidolin D, Tortorella C, De Caro R, Agnati LF. A Self-Similarity Logic May Shape the Organization of the Nervous System. ADVANCES IN NEUROBIOLOGY 2024; 36:203-225. [PMID: 38468034 DOI: 10.1007/978-3-031-47606-8_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Abstract
From the morphological point of view, the nervous system exhibits a fractal, self-similar geometry at various levels of observations, from single cells up to cell networks. From the functional point of view, it is characterized by a hierarchical organization in which self-similar structures (networks) of different miniaturizations are nested within each other. In particular, neuronal networks, interconnected to form neuronal systems, are formed by neurons, which operate thanks to their molecular networks, mainly having proteins as components that via protein-protein interactions can be assembled in multimeric complexes working as micro-devices. On this basis, the term "self-similarity logic" was introduced to describe a nested organization where, at the various levels, almost the same rules (logic) to perform operations are used. Self-similarity and self-similarity logic both appear to be intimately linked to the biophysical evidence for the nervous system being a pattern-forming system that can flexibly switch from one coherent state to another. Thus, they can represent the key concepts to describe its complexity and its concerted, holistic behavior.
Collapse
Affiliation(s)
- Diego Guidolin
- Department of Neuroscience, University of Padova, Padova, Italy.
| | | | | | - Luigi F Agnati
- Department of Biomedical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
4
|
Xiao M, Hou J, Xu M, Li S, Yang B. Aquaporins in Nervous System. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1398:99-124. [PMID: 36717489 DOI: 10.1007/978-981-19-7415-1_7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Aquaporins (AQPs) mediate water flux between the four distinct water compartments in the central nervous system (CNS). In the present chapter, we mainly focus on the expression and function of the nine AQPs expressed in the CNS, which include five members of aquaporin subfamily: AQP1, AQP4, AQP5, AQP6, and AQP8; three members of aquaglyceroporin subfamily: AQP3, AQP7, and AQP9; and one member of superaquaporin subfamily: AQP11. In addition, AQP1, AQP2, and AQP4 expressed in the peripheral nervous system are also reviewed. AQP4, the predominant water channel in the CNS, is involved both in the astrocyte swelling of cytotoxic edema and the resolution of vasogenic edema and is of pivotal importance in the pathology of brain disorders such as neuromyelitis optica, brain tumors, and neurodegenerative disorders. Moreover, AQP4 has been demonstrated as a functional regulator of recently discovered glymphatic system that is a main contributor to clearance of toxic macromolecule from the brain. Other AQPs are also involved in a variety of important physiological and pathological process in the brain. It has been suggested that AQPs could represent an important target in treatment of brain disorders like cerebral edema. Future investigations are necessary to elucidate the pathological significance of AQPs in the CNS.
Collapse
Affiliation(s)
- Ming Xiao
- Jiangsu Province, Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, China
| | - Jiaoyu Hou
- Department of Geriatrics, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Mengmeng Xu
- Basic Medical College, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Shao Li
- Department of Physiology, Dalian Medical University, Dalian, China
| | - Baoxue Yang
- School of Basic Medical Sciences, Peking University, Beijing, China.
| |
Collapse
|
5
|
Carvalho GB, Damasio A. Interoception and the origin of feelings: A new synthesis. Bioessays 2021; 43:e2000261. [PMID: 33763881 DOI: 10.1002/bies.202000261] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 02/26/2021] [Accepted: 03/02/2021] [Indexed: 02/06/2023]
Abstract
Feelings are conscious mental events that represent body states as they undergo homeostatic regulation. Feelings depend on the interoceptive nervous system (INS), a collection of peripheral and central pathways, nuclei and cortical regions which continuously sense chemical and anatomical changes in the organism. How such humoral and neural signals come to generate conscious mental states has been a major scientific question. The answer proposed here invokes (1) several distinctive and poorly known physiological features of the INS; and (2) a unique interaction between the body (the 'object' of interoception) and the central nervous system (which generates the 'subject' of interoception). The atypical traits of the INS and the direct interactions between neural and non-neural physiological compartments of the organism, neither of which is present in exteroceptive systems, plausibly explain the qualitative and subjective aspects of feelings, thus accounting for their conscious nature.
Collapse
Affiliation(s)
- Gil B Carvalho
- Brain and Creativity Institute, Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, California, USA
| | - Antonio Damasio
- Brain and Creativity Institute, Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
6
|
Abstract
Opioids are the most powerful analgesics available to date. However, they may also induce adverse effects including paradoxical opioid-induced hyperalgesia. A mechanism that might underlie opioid-induced hyperalgesia is the amplification of synaptic strength at spinal C-fibre synapses after withdrawal from systemic opioids such as remifentanil ("opioid-withdrawal long-term potentiation [LTP]"). Here, we show that both the induction as well as the maintenance of opioid-withdrawal LTP were abolished by pharmacological blockade of spinal glial cells. By contrast, the blockade of TLR4 had no effect on the induction of opioid-withdrawal LTP. D-serine, which may be released upon glial cell activation, was necessary for withdrawal LTP. D-serine is the dominant coagonist for neuronal NMDA receptors, which are required for the amplification of synaptic strength on remifentanil withdrawal. Unexpectedly, opioid-withdrawal LTP was transferable through the cerebrospinal fluid between animals. This suggests that glial-cell-derived mediators accumulate in the extracellular space and reach the cerebrospinal fluid at biologically active concentrations, thereby creating a soluble memory trace that is transferable to another animal ("transfer LTP"). When we enzymatically degraded D-serine in the superfusate, LTP could no longer be transferred. Transfer LTP was insensitive to pharmacological blockade of glial cells in the recipient animal, thus representing a rare form of glial cell-independent LTP in the spinal cord.
Collapse
|
7
|
Kempe PRG, Chiarotto GB, Barraviera B, Ferreira RS, de Oliveira ALR. Neuroprotection and immunomodulation by dimethyl fumarate and a heterologous fibrin biopolymer after ventral root avulsion and reimplantation. J Venom Anim Toxins Incl Trop Dis 2020; 26:e20190093. [PMID: 32518556 PMCID: PMC7250131 DOI: 10.1590/1678-9199-jvatitd-2019-0093] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Background: Ventral root avulsion (VRA) is an experimental approach in which there is an abrupt separation of the motor roots from the surface of the spinal cord. As a result, most of the axotomized motoneurons degenerate by the second week after injury, and the significant loss of synapses and increased glial reaction triggers a chronic inflammatory state. Pharmacological treatment associated with root reimplantation is thought to overcome the degenerative effects of VRA. Therefore, treatment with dimethyl fumarate (DMF), a drug with neuroprotective and immunomodulatory effects, in combination with a heterologous fibrin sealant/biopolymer (FS), a biological glue, may improve the regenerative response. Methods: Adult female Lewis rats were subjected to VRA of L4-L6 roots followed by reimplantation and daily treatment with DMF for four weeks. Survival times were evaluated 1, 4 or 12 weeks after surgery. Neuronal survival assessed by Nissl staining, glial reactivity (anti-GFAP for astrocytes and anti-Iba-1 for microglia) and synapse preservation (anti-VGLUT1 for glutamatergic inputs and anti-GAD65 for GABAergic inputs) evaluated by immunofluorescence, gene expression (pro- and anti-inflammatory molecules) and motor function recovery were measured. Results: Treatment with DMF at a dose of 15 mg/kg was found to be neuroprotective and immunomodulatory because it preserved motoneurons and synapses and decreased astrogliosis and microglial reactions, as well as downregulated the expression of pro-inflammatory gene transcripts. Conclusion: The pharmacological benefit was further enhanced when associated with root reimplantation with FS, in which animals recovered at least 50% of motor function, showing the efficacy of employing multiple regenerative approaches following spinal cord root injury.
Collapse
Affiliation(s)
- Paula R G Kempe
- Laboratory of Nerve Regeneration, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | | | - Benedito Barraviera
- Center for the Study of Venoms and Venomous Animals (CEVAP), São Paulo State University (UNESP), Botucatu, SP, Brazil
| | - Rui Seabra Ferreira
- Center for the Study of Venoms and Venomous Animals (CEVAP), São Paulo State University (UNESP), Botucatu, SP, Brazil
| | | |
Collapse
|
8
|
Capo-Rangel G, Gerardo-Giorda L, Somersalo E, Calvetti D. Metabolism plays a central role in the cortical spreading depression: Evidence from a mathematical model. J Theor Biol 2020; 486:110093. [PMID: 31778711 DOI: 10.1016/j.jtbi.2019.110093] [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: 08/01/2019] [Revised: 10/28/2019] [Accepted: 11/23/2019] [Indexed: 11/24/2022]
Abstract
The slow propagating waves of strong depolarization of neural cells characterizing cortical spreading depression, or depolarization, (SD) are known to break cerebral homeostasis and induce significant hemodynamic and electro-metabolic alterations. Mathematical models of cortical spreading depression found in the literature tend to focus on the changes occurring at the electrophysiological level rather than on the ensuing metabolic changes. In this paper, we propose a novel mathematical model which is able to simulate the coupled electrophysiology and metabolism dynamics of SD events, including the swelling of neurons and astrocytes and the concomitant shrinkage of extracellular space. The simulations show that the metabolic coupling leads to spontaneous repetitions of the SD events, which the electrophysiological model alone is not capable to produce. The model predictions, which corroborate experimental findings from the literature, show a strong disruption in metabolism accompanying each wave of spreading depression in the form of a sharp decrease of glucose and oxygen concentrations, with a simultaneous increase in lactate concentration which, in turn, delays the clearing of excess potassium in extracellular space. Our model suggests that the depletion of glucose and oxygen concentration is more pronounced in astrocyte than neuron, in line with the partitioning of the energetic cost of potassium clearing. The model suggests that the repeated SD events are electro-metabolic oscillations that cannot be explained by the electrophysiology alone. The model highlights the crucial role of astrocytes in cleaning the excess potassium flooding extracellular space during a spreading depression event: further, if the ratio of glial/neuron density increases, the frequency of cortical SD events decreases, and the peak potassium concentration in extracellular space is lower than with equal volume fractions.
Collapse
Affiliation(s)
| | | | - E Somersalo
- Basque Center for Applied Mathematics, Spain
| | - D Calvetti
- Department of Mathematics, Applied Mathematics and Statistics, Case Western Reserve University, Ohio.
| |
Collapse
|
9
|
A Role for The P2Y1 Receptor in Nonsynaptic Cross-depolarization in the Rat Dorsal Root Ganglia. Neuroscience 2019; 423:98-108. [PMID: 31689490 DOI: 10.1016/j.neuroscience.2019.09.038] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 09/19/2019] [Accepted: 09/27/2019] [Indexed: 12/19/2022]
Abstract
Non-synaptic transmission is pervasive throughout the nervous system. It appears especially prevalent in peripheral ganglia, where non-synaptic interactions between neighboring cell bodies have been described in both physiological and pathological conditions, a phenomenon referred to as cross-depolarization (CD) and thought to play a role in sensory processing and chronic pain. CD has been proposed to be mediated by a chemical agent, but its identity has remained elusive. Here, we report that in the rat dorsal root ganglion (DRG), the P2Y1 purinergic receptor (P2RY1) plays an important role in regulating CD. The effect of P2RY1 is cell-type specific: pharmacological blockade of P2RY1 inhibited CD in A-type neurons while enhancing it in C-type neurons. In the nodose ganglion of the vagus, CD requires extracellular calcium in a large percentage of cells. In contrast, we show that in the DRG extracellular calcium appears to play no major role, pointing to a mechanistic difference between the two peripheral ganglia. Furthermore, we show that DRG glial cells also play a cell-type specific role in CD regulation. Fluorocitrate-induced glial inactivation had no effect on A-cells but enhanced CD in C-cells. These findings shed light on the mechanism of CD in the DRG and pave the way for further analysis of non-synaptic neuronal communication in sensory ganglia.
Collapse
|
10
|
Guidolin D, Marcoli M, Tortorella C, Maura G, Agnati LF. From the hierarchical organization of the central nervous system to the hierarchical aspects of biocodes. Biosystems 2019; 183:103975. [PMID: 31128147 DOI: 10.1016/j.biosystems.2019.103975] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 05/21/2019] [Accepted: 05/22/2019] [Indexed: 12/17/2022]
Abstract
The quite recent (at least on the evolutionary time scale) emergence of nervous systems in complex organisms enabled the living beings to build a wide-ranging model of the external world in order to predict and evaluate the outcomes of their actions. Such a process likely represents a real coding activity, since, by proper handling of information, it generates a mapping between the external environment and internal cerebral activity patterns. The patterns of neural activity that correspond to the final maps, however, emerge from the holistic assembly of a multilevel functional organization. Nerve tissue components, indeed, appear organized in compartments, also called functional modules (FM), that contain system components and circuits of different miniaturizations not only arranged to work together either in parallel or in series but also nested within each other. At least three levels can be recognized in a functional module and it is possible to point out that such a hierarchical organization of the brain circuits could be mirrored by a corresponding hierarchical organization of biocodes. This feature can also suggest the hypothesis that the same logic could operate also at system level to integrate FM into functional brain areas and to associate areas to generate the final map used by humans to image the external world and to imagine untestable worlds.
Collapse
Affiliation(s)
- D Guidolin
- Department of Neuroscience, Section of Anatomy, University of Padova, via Gabelli 65, 35121 Padova, Italy.
| | - M Marcoli
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Viale Cembrano 4, 16148, Genova, Italy
| | - C Tortorella
- Department of Neuroscience, Section of Anatomy, University of Padova, via Gabelli 65, 35121 Padova, Italy
| | - G Maura
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Viale Cembrano 4, 16148, Genova, Italy
| | - L F Agnati
- Department of Diagnostic, Clinical Medicine and Public Health, University of Modena and Reggio Emilia, Via Campi 287, 41125, Modena, Italy; Department of Neuroscience, Karolinska Institutet, Retzius väg 8, Stockholm, Sweden
| |
Collapse
|
11
|
Agnati LF, Marcoli M, Maura G, Woods A, Guidolin D. The brain as a "hyper-network": the key role of neural networks as main producers of the integrated brain actions especially via the "broadcasted" neuroconnectomics. J Neural Transm (Vienna) 2018; 125:883-897. [PMID: 29427068 DOI: 10.1007/s00702-018-1855-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 02/04/2018] [Indexed: 02/07/2023]
Abstract
Investigations of brain complex integrative actions should consider beside neural networks, glial, extracellular molecular, and fluid channels networks. The present paper proposes that all these networks are assembled into the brain hyper-network that has as fundamental components, the tetra-partite synapses, formed by neural, glial, and extracellular molecular networks. Furthermore, peri-synaptic astrocytic processes by modulating the perviousness of extracellular fluid channels control the signals impinging on the tetra-partite synapses. It has also been surmised that global signalling via astrocytes networks and highly pervasive signals, such as electromagnetic fields (EMFs), allow the appropriate integration of the various networks especially at crucial nodes level, the tetra-partite synapses. As a matter of fact, it has been shown that astrocytes can form gap-junction-coupled syncytia allowing intercellular communication characterised by a rapid and possibly long-distance transfer of signals. As far as the EMFs are concerned, the concept of broadcasted neuroconnectomics (BNC) has been introduced to describe highly pervasive signals involved in resetting the information handling of brain networks at various miniaturisation levels. In other words, BNC creates, thanks to the EMFs, generated especially by neurons, different assemblages among the various networks forming the brain hyper-network. Thus, it is surmised that neuronal networks are the "core components" of the brain hyper-network that has as special "nodes" the multi-facet tetra-partite synapses. Furthermore, it is suggested that investigations on the functional plasticity of multi-partite synapses in response to BNC can be the background for a new understanding and perhaps a new modelling of brain morpho-functional organisation and integrative actions.
Collapse
Affiliation(s)
- Luigi F Agnati
- Department of Diagnostic, Clinical Medicine and Public Health, University of Modena and Reggio Emilia, Modena, Italy. .,Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| | - Manuela Marcoli
- Section of Pharmacology and Toxicology, Department of Pharmacy, University of Genova, Viale Cembrano 4, 16148, Genoa, Italy. .,Centre of Excellence for Biomedical Research CEBR, University of Genova, Genoa, Italy.
| | - Guido Maura
- Section of Pharmacology and Toxicology, Department of Pharmacy, University of Genova, Viale Cembrano 4, 16148, Genoa, Italy
| | - Amina Woods
- Structural Biology Unit, National Institutes of Health, National Institute of Drug Abuse-Intramural Research Program, Baltimore, MD, 21224, USA
| | - Diego Guidolin
- Department of Molecular Medicine, University of Padova, Padua, Italy
| |
Collapse
|
12
|
Guidolin D, Marcoli M, Maura G, Agnati LF. New dimensions of connectomics and network plasticity in the central nervous system. Rev Neurosci 2018; 28:113-132. [PMID: 28030363 DOI: 10.1515/revneuro-2016-0051] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 09/20/2016] [Indexed: 12/24/2022]
Abstract
Cellular network architecture plays a crucial role as the structural substrate for the brain functions. Therefore, it represents the main rationale for the emerging field of connectomics, defined as the comprehensive study of all aspects of central nervous system connectivity. Accordingly, in the present paper the main emphasis will be on the communication processes in the brain, namely wiring transmission (WT), i.e. the mapping of the communication channels made by cell components such as axons and synapses, and volume transmission (VT), i.e. the chemical signal diffusion along the interstitial brain fluid pathways. Considering both processes can further expand the connectomics concept, since both WT-connectomics and VT-connectomics contribute to the structure of the brain connectome. A consensus exists that such a structure follows a hierarchical or nested architecture, and macro-, meso- and microscales have been defined. In this respect, however, several lines of evidence indicate that a nanoscale (nano-connectomics) should also be considered to capture direct protein-protein allosteric interactions such as those occurring, for example, in receptor-receptor interactions at the plasma membrane level. In addition, emerging evidence points to novel mechanisms likely playing a significant role in the modulation of intercellular connectivity, increasing the plasticity of the system and adding complexity to its structure. In particular, the roamer type of VT (i.e. the intercellular transfer of RNA, proteins and receptors by extracellular vesicles) will be discussed since it allowed us to introduce a new concept of 'transient changes of cell phenotype', that is the transient acquisition of new signal release capabilities and/or new recognition/decoding apparatuses.
Collapse
|
13
|
Stokum JA, Kwon MS, Woo SK, Tsymbalyuk O, Vennekens R, Gerzanich V, Simard JM. SUR1-TRPM4 and AQP4 form a heteromultimeric complex that amplifies ion/water osmotic coupling and drives astrocyte swelling. Glia 2017; 66:108-125. [PMID: 28906027 DOI: 10.1002/glia.23231] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 07/23/2017] [Accepted: 08/23/2017] [Indexed: 12/17/2022]
Abstract
Astrocyte swelling occurs after central nervous system injury and contributes to brain swelling, which can increase mortality. Mechanisms proffered to explain astrocyte swelling emphasize the importance of either aquaporin-4 (AQP4), an astrocyte water channel, or of Na+ -permeable channels, which mediate cellular osmolyte influx. However, the spatio-temporal functional interactions between AQP4 and Na+ -permeable channels that drive swelling are poorly understood. We hypothesized that astrocyte swelling after injury is linked to an interaction between AQP4 and Na+ -permeable channels that are newly upregulated. Here, using co-immunoprecipitation and Förster resonance energy transfer, we report that AQP4 physically co-assembles with the sulfonylurea receptor 1-transient receptor potential melastatin 4 (SUR1-TRPM4) monovalent cation channel to form a novel heteromultimeric water/ion channel complex. In vitro cell-swelling studies using calcein fluorescence imaging of COS-7 cells expressing various combinations of AQP4, SUR1, and TRPM4 showed that the full tripartite complex, comprised of SUR1-TRPM4-AQP4, was required for fast, high-capacity transmembrane water transport that drives cell swelling, with these findings corroborated in cultured primary astrocytes. In a murine model of brain edema involving cold-injury to the cerebellum, we found that astrocytes newly upregulate SUR1-TRPM4, that AQP4 co-associates with SUR1-TRPM4, and that genetic inactivation of the solute pore of the SUR1-TRPM4-AQP4 complex blocked in vivo astrocyte swelling measured by diolistic labeling, thereby corroborating our in vitro functional studies. Together, these findings demonstrate a novel molecular mechanism involving the SUR1-TRPM4-AQP4 complex to account for bulk water influx during astrocyte swelling. These findings have broad implications for the understanding and treatment of AQP4-mediated pathological conditions.
Collapse
Affiliation(s)
- Jesse A Stokum
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland, 21201-1595
| | - Min S Kwon
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland, 21201-1595
| | - Seung K Woo
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland, 21201-1595
| | - Orest Tsymbalyuk
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland, 21201-1595
| | - Rudi Vennekens
- Department of Cellular and Molecular Medicine, Laboratory of Ion Channel Research, Katholieke Universiteit Leuven, Leuven, 3000, Belgium
| | - Volodymyr Gerzanich
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland, 21201-1595
| | - J Marc Simard
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland, 21201-1595.,Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland, 21201-1595.,Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, 21201-1595
| |
Collapse
|
14
|
Abstract
Aquaporins (AQPs ) mediate water flux between the four distinct water compartments in the central nervous system (CNS). In the present chapter, we mainly focus on the expression and function of the 9 AQPs expressed in the CNS, which include five members of aquaporin subfamily: AQP1, AQP4, AQP5, AQP6, and AQP8; three members of aquaglyceroporin subfamily: AQP3, AQP7, and AQP9; and one member of superaquaporin subfamily: AQP11. In addition, AQP1, AQP2 and AQP4 expressed in the peripheral nervous system (PNS) are also reviewed. AQP4, the predominant water channel in the CNS, is involved both in the astrocyte swelling of cytotoxic edema and the resolution of vasogenic edema, and is of pivotal importance in the pathology of brain disorders such as neuromyelitis optica , brain tumors and Alzheimer's disease. Other AQPs are also involved in a variety of important physiological and pathological process in the brain. It has been suggested that AQPs could represent an important target in treatment of brain disorders like cerebral edema. Future investigations are necessary to elucidate the pathological significance of AQPs in the CNS.
Collapse
|
15
|
Arango-Dávila CA, Rincón-Hoyos HG. Depressive Disorder, Anxiety Disorder and Chronic Pain: Multiple Manifestations of a Common Clinical and Pathophysiological Core. ACTA ACUST UNITED AC 2016; 47:46-55. [PMID: 29428122 DOI: 10.1016/j.rcp.2016.10.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Revised: 10/10/2016] [Accepted: 10/31/2016] [Indexed: 02/03/2023]
Abstract
INTRODUCTION A high proportion of depressive disorders are accompanied by anxious manifestations, just as depression and anxiety often present with many painful manifestations, or conversely, painful manifestations cause or worsen depressive and anxious expressions. There is increasingly more evidence of the pathophysiological, and neurophysiological and technical imaging similarity of pain and depression. METHODS Narrative review of the pathophysiological and clinical aspects of depression and chronic pain comorbidity. Research articles are included that emphasise the most relevant elements related to understanding the pathophysiology of both manifestations. RESULTS The pathological origin, physiology and clinical approach to these disorders have been more clearly established with the latest advances in biochemical and cellular techniques, as well as the advent of imaging technologies. This information is systematised with comprehensive images and clinical pictures. CONCLUSIONS The recognition that the polymorphism of inflammation-related genes generates susceptibility to depressive manifestations and may modify the response to antidepressant treatments establishes that the inflammatory response is not only an aetiopathogenic component of pain, but also of stress and depression. Likewise, the similarity in approach with images corroborates not only the structural, but the functional and pathophysiological analogy between depression and chronic pain. Knowledge of depression-anxiety-chronic pain comorbidity is essential in the search for effective therapeutic interventions.
Collapse
Affiliation(s)
- Cesar A Arango-Dávila
- Sección de Psiquiatría y Psicología, Fundación Valle del Lili, Cali, Colombia; Grupo de Investigación Biomédica, Universidad Icesi, Cali, Colombia.
| | - Hernán G Rincón-Hoyos
- Sección de Psiquiatría y Psicología, Fundación Valle del Lili, Cali, Colombia; Department of Psychiatry and Behavioral Sciences, Leonard M. Miller School of Medicine, University of Miami, Miami, Estados Unidos
| |
Collapse
|
16
|
Minieri L, Pivonkova H, Harantova L, Anderova M, Ferroni S. Intracellular Na+
inhibits volume-regulated anion channel in rat cortical astrocytes. J Neurochem 2015; 132:286-300. [DOI: 10.1111/jnc.12962] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Revised: 09/04/2014] [Accepted: 09/25/2014] [Indexed: 01/08/2023]
Affiliation(s)
- Laura Minieri
- Department of Pharmacy and Biotechnology; University of Bologna; Bologna Italy
| | - Helena Pivonkova
- Department of Cellular Neurophysiology; Institute of Experimental Medicine; Academy of Sciences of the Czech Republic; Prague Czech Republic
| | - Lenka Harantova
- Department of Cellular Neurophysiology; Institute of Experimental Medicine; Academy of Sciences of the Czech Republic; Prague Czech Republic
| | - Miroslava Anderova
- Department of Cellular Neurophysiology; Institute of Experimental Medicine; Academy of Sciences of the Czech Republic; Prague Czech Republic
| | - Stefano Ferroni
- Department of Pharmacy and Biotechnology; University of Bologna; Bologna Italy
| |
Collapse
|
17
|
Henckens MJAG, van der Marel K, van der Toorn A, Pillai AG, Fernández G, Dijkhuizen RM, Joëls M. Stress-induced alterations in large-scale functional networks of the rodent brain. Neuroimage 2014; 105:312-22. [PMID: 25462693 DOI: 10.1016/j.neuroimage.2014.10.037] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Revised: 09/20/2014] [Accepted: 10/08/2014] [Indexed: 02/07/2023] Open
Abstract
Stress-related psychopathology is associated with altered functioning of large-scale brain networks. Animal research into chronic stress, one of the most prominent environmental risk factors for development of psychopathology, has revealed molecular and cellular mechanisms potentially contributing to human mental disease. However, so far, these studies have not addressed the system-level changes in extended brain networks, thought to critically contribute to mental disorders. We here tested the effects of chronic stress exposure (10 days immobilization) on the structural integrity and functional connectivity patterns in the brain, using high-resolution structural MRI, diffusion kurtosis imaging, and resting-state functional MRI, while confirming the expected changes in neuronal dendritic morphology using Golgi-staining. Stress effectiveness was confirmed by a significantly lower body weight and increased adrenal weight. In line with previous research, stressed animals displayed neuronal dendritic hypertrophy in the amygdala and hypotrophy in the hippocampal and medial prefrontal cortex. Using independent component analysis of resting-state fMRI data, we identified ten functional connectivity networks in the rodent brain. Chronic stress appeared to increase connectivity within the somatosensory, visual, and default mode networks. Moreover, chronic stress exposure was associated with an increased volume and diffusivity of the lateral ventricles, whereas no other volumetric changes were observed. This study shows that chronic stress exposure in rodents induces alterations in functional network connectivity strength which partly resemble those observed in stress-related psychopathology. Moreover, these functional consequences of stress seem to be more prominent than the effects on gross volumetric change, indicating their significance for future research.
Collapse
Affiliation(s)
- Marloes J A G Henckens
- Department of Neuroscience and Pharmacology, Rudolf Magnus Institute of Neuroscience, University Medical Center Utrecht, 3584 CG Utrecht, The Netherlands; Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen, 6500 HB Nijmegen, The Netherlands.
| | - Kajo van der Marel
- Biomedical MR Imaging and Spectroscopy Group, Image Sciences Institute, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Annette van der Toorn
- Biomedical MR Imaging and Spectroscopy Group, Image Sciences Institute, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Anup G Pillai
- Department of Neuroscience and Pharmacology, Rudolf Magnus Institute of Neuroscience, University Medical Center Utrecht, 3584 CG Utrecht, The Netherlands
| | - Guillén Fernández
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen, 6500 HB Nijmegen, The Netherlands; Department of Cognitive Neuroscience, Radboud University Medical Centre, 6500 HB Nijmegen, The Netherlands
| | - Rick M Dijkhuizen
- Biomedical MR Imaging and Spectroscopy Group, Image Sciences Institute, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Marian Joëls
- Department of Neuroscience and Pharmacology, Rudolf Magnus Institute of Neuroscience, University Medical Center Utrecht, 3584 CG Utrecht, The Netherlands
| |
Collapse
|
18
|
Florence CM, Baillie LD, Mulligan SJ. Dynamic volume changes in astrocytes are an intrinsic phenomenon mediated by bicarbonate ion flux. PLoS One 2012; 7:e51124. [PMID: 23226475 PMCID: PMC3511399 DOI: 10.1371/journal.pone.0051124] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Accepted: 10/30/2012] [Indexed: 11/18/2022] Open
Abstract
Astrocytes, the major type of non-neuronal cells in the brain, play an important functional role in extracellular potassium ([K(+)](o)) and pH homeostasis. Pathological brain states that result in [K(+)](o) and pH dysregulation have been shown to cause astrocyte swelling. However, whether astrocyte volume changes occur under physiological conditions is not known. In this study we used two-photon imaging to visualize real-time astrocyte volume changes in the stratum radiatum of the hippocampus CA1 region. Astrocytes were observed to swell by 19.0±0.9% in response to a small physiological increase in the concentration of [K(+)](o) (3 mM). Astrocyte swelling was mediated by the influx of bicarbonate (HCO(3-)) ions as swelling was significantly decreased when the influx of HCO(3-) was reduced. We found: 1) in HCO(3-) free extracellular solution astrocytes swelled by 5.4±0.7%, 2) when the activity of the sodium-bicarbonate cotransporter (NBC) was blocked the astrocytes swelled by 8.3±0.7%, and 3) in the presence of an extracellular carbonic anhydrase (CA) inhibitor astrocytes swelled by 11.4±0.6%. Because a significant HCO(3-) efflux is known to occur through the γ-amino-butyric acid (GABA) channel, we performed a series of experiments to determine if astrocytes were capable of HCO(3-) mediated volume shrinkage with GABA channel activation. Astrocytes were found to shrink -7.7±0.5% of control in response to the GABA(A) channel agonist muscimol. Astrocyte shrinkage from GABA(A) channel activation was significantly decreased to -5.0±0.6% of control in the presence of the membrane-permeant CA inhibitor acetazolamide (ACTZ). These dynamic astrocyte volume changes may represent a previously unappreciated yet fundamental mechanism by which astrocytes regulate physiological brain functioning.
Collapse
Affiliation(s)
- Clare M. Florence
- Department of Physiology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Landon D. Baillie
- Department of Physiology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Sean J. Mulligan
- Department of Physiology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| |
Collapse
|
19
|
Lanciotti A, Brignone MS, Molinari P, Visentin S, De Nuccio C, Macchia G, Aiello C, Bertini E, Aloisi F, Petrucci TC, Ambrosini E. Megalencephalic leukoencephalopathy with subcortical cysts protein 1 functionally cooperates with the TRPV4 cation channel to activate the response of astrocytes to osmotic stress: dysregulation by pathological mutations. Hum Mol Genet 2012; 21:2166-80. [PMID: 22328087 DOI: 10.1093/hmg/dds032] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Megalencephalic leukoencephalopathy with subcortical cysts (MLC), a rare leukodystrophy characterized by macrocephaly, subcortical fluid cysts and myelin vacuolation, has been linked to mutations in the MLC1 gene. This gene encodes a membrane protein that is highly expressed in astrocytes. Based on MLC pathological features, it was proposed that astrocyte-mediated defects in ion and fluid homeostasis could account for the alterations observed in MLC-affected brains. However, the role of MLC1 and the effects of pathological mutations on astrocyte osmoregulatory functions have still to be demonstrated. Using human astrocytoma cells stably overexpressing wild-type MLC1 or three known MLC-associated pathological mutations, we investigated MLC1 involvement in astrocyte reaction to osmotic changes using biochemical, dynamic video imaging and immunofluorescence techniques. We have found that MLC1 overexpressed in astrocytoma cells is mainly localized in the plasma membrane, is part of the Na,K-ATPase-associated molecular complex that includes the potassium channel Kir4.1, syntrophin and aquaporin-4 and functionally interacts with the calcium permeable channel TRPV4 (transient receptor potential vanilloid-4 cation channel) which mediates swelling-induced cytosolic calcium increase and volume recovery in response to hyposmosis. Pathological MLC mutations cause changes in MLC1 expression and intracellular localization as well as in the astrocyte response to osmotic changes by altering MLC1 molecular interactions with the Na,K-ATPase molecular complex and abolishing the increase in calcium influx induced by hyposmosis and treatment with the TRPV4 agonist 4αPDD. These data demonstrate, for the first time, that MLC1 plays a role in astrocyte osmo-homeostasis and that defects in intracellular calcium dynamics may contribute to MLC pathogenesis.
Collapse
Affiliation(s)
- Angela Lanciotti
- Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità,Viale Regina Elena 299, Rome 00161, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Iacovetta C, Rudloff E, Kirby R. The role of aquaporin 4 in the brain. Vet Clin Pathol 2012; 41:32-44. [PMID: 22250904 DOI: 10.1111/j.1939-165x.2011.00390.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2011] [Revised: 11/14/2011] [Accepted: 12/10/2011] [Indexed: 11/26/2022]
Abstract
Emerging evidence suggests that aquaporin (AQP) 4 water channels play an important role in water homeostasis in the brain. These water channels are most abundant in the cell membrane of astrocytes, but are also present within ependymal cell membranes and in osmosensory areas of the hypothalamus. Water transport through AQP4 depends on concentration gradients across the membrane, but the rate of transport is determined by the capacity of astrocytes to up- and down-regulate AQP4 numbers, their location within the membrane, and the overall permeability of the channel. Other functions of brain AQP4 involve potassium uptake and release by astrocytes, migration of glial cells, glial scarring, and astrocyte-to-astrocyte cell communication. AQP water channels are involved in formation and control of edema in the brain and in multiple disease processes in the brain, such as seizures and tumors. There is abundant scientific literature on AQP4 describing its structure, function, location, and role in water homeostasis and edema in the brain. Investigation of AQP expression in the canine and feline brain should be pursued so that clinically relevant comparisons between findings in mice, rats, and people and animal patients can be made.
Collapse
|
21
|
Guidolin D, Albertin G, Guescini M, Fuxe K, Agnati L. Central Nervous System and Computation. QUARTERLY REVIEW OF BIOLOGY 2011; 86:265-85. [DOI: 10.1086/662456] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
|
22
|
Adermark L, Clarke RBC, Olsson T, Hansson E, Söderpalm B, Ericson M. Implications for glycine receptors and astrocytes in ethanol-induced elevation of dopamine levels in the nucleus accumbens. Addict Biol 2011; 16:43-54. [PMID: 20331561 DOI: 10.1111/j.1369-1600.2010.00206.x] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Elevated dopamine levels are believed to contribute to the rewarding sensation of ethanol (EtOH), and previous research has shown that strychnine-sensitive glycine receptors in the nucleus accumbens (nAc) are involved in regulating dopamine release and in mediating the reinforcing effects of EtOH. Furthermore, the osmoregulator taurine, which is released from astrocytes treated with EtOH, can act as an endogenous ligand for the glycine receptor, and increase extracellular dopamine levels. The aim of this study was to address if EtOH-induced swelling of astrocytes could contribute to elevated dopamine levels by increasing the extracellular concentration of taurine. Cell swelling was estimated by optical sectioning of fluorescently labeled astrocytes in primary cultures from rat, and showed that EtOH (25-150 mM) increased astrocyte cell volumes in a concentration- and ion-dependent manner. The EtOH-induced cell swelling was inhibited in cultures treated with the Na(+) /K(+) /2Cl⁻ cotransporter blocker furosemide (1 mM), Na(+) /K(+) -ATPase inhibitor ouabain (0.1 mM), potassium channel inhibitor BaCl₂ (50 µM) and in cultures containing low extracellular sodium concentration (3 mM). In vivo microdialysis performed in the nAc of awake and freely moving rats showed that local treatment with EtOH enhanced the concentrations of dopamine and taurine in the microdialysate, while glycine and β-alanine levels were not significantly modulated. EtOH-induced dopamine release was antagonized by local treatment with the glycine receptor antagonist strychnine (20 µM) or furosemide (100 µM or 1 mM). Furosemide also prevented EtOH-induced taurine release in the nAc. In conclusion, our data suggest that extracellular concentrations of dopamine and taurine are interconnected and that swelling of astrocytes contributes to the acute rewarding sensation of EtOH.
Collapse
Affiliation(s)
- Louise Adermark
- Department of Psychiatry and Neurochemistry, Institute for Neuroscience and Physiology, University of Gothenburg, Sweden.
| | | | | | | | | | | |
Collapse
|
23
|
|
24
|
Benfenati V, Ferroni S. Water transport between CNS compartments: functional and molecular interactions between aquaporins and ion channels. Neuroscience 2009; 168:926-40. [PMID: 20026249 DOI: 10.1016/j.neuroscience.2009.12.017] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2009] [Revised: 12/04/2009] [Accepted: 12/05/2009] [Indexed: 12/16/2022]
Abstract
The physiological ability of the mammalian CNS to integrate peripheral stimuli and to convey information to the body is tightly regulated by its capacity to preserve the ion composition and volume of the perineuronal milieu. It is well known that astroglial syncytium plays a crucial role in such process by controlling the homeostasis of ions and water through the selective transmembrane movement of inorganic and organic molecules and the equilibration of osmotic gradients. Astrocytes, in fact, by contacting neurons and cells lining the fluid-filled compartments, are in a strategic position to fulfill this role. They are endowed with ion and water channel proteins that are localized in specific plasma membrane domains facing diverse liquid spaces. Recent data in rodents have demonstrated that the precise dynamics of the astroglia-mediated homeostatic regulation of the CNS is dependent on the interactions between water channels and ion channels, and their anchoring with proteins that allow the formation of macromolecular complexes in specific cellular domains. Interplay can occur with or without direct molecular interactions suggesting the existence of different regulatory mechanisms. The importance of molecular and functional interactions is pinpointed by the numerous observations that as consequence of pathological insults leading to the derangement of ion and volume homeostasis the cell surface expression and/or polarized localization of these proteins is perturbed. Here, we critically discuss the experimental evidence concerning: (1) molecular and functional interplay of aquaporin 4, the major aquaporin protein in astroglial cells, with potassium and gap-junctional channels that are involved in extracellular potassium buffering. (2) the interactions of aquaporin 4 with chloride and calcium channels regulating cell volume homeostasis. The relevance of the crosstalk between water channels and ion channels in the pathogenesis of astroglia-related acute and chronic diseases of the CNS is also briefly discussed.
Collapse
Affiliation(s)
- V Benfenati
- Istituto per lo Studio dei Materiali Nanostrutturati, ISMN, National Research Council, Via Gobetti 101, 40129 Bologna, Italy
| | | |
Collapse
|
25
|
Mandl RCW, Schnack HG, Zwiers MP, van der Schaaf A, Kahn RS, Hulshoff Pol HE. Functional diffusion tensor imaging: measuring task-related fractional anisotropy changes in the human brain along white matter tracts. PLoS One 2008; 3:e3631. [PMID: 18982065 PMCID: PMC2574009 DOI: 10.1371/journal.pone.0003631] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2008] [Accepted: 10/10/2008] [Indexed: 11/19/2022] Open
Abstract
Background Functional neural networks in the human brain can be studied from correlations between activated gray matter regions measured with fMRI. However, while providing important information on gray matter activation, no information is gathered on the co-activity along white matter tracts in neural networks. Methodology/Principal Findings We report on a functional diffusion tensor imaging (fDTI) method that measures task-related changes in fractional anisotropy (FA) along white matter tracts. We hypothesize that these fractional anisotropy changes relate to morphological changes of glial cells induced by axonal activity although the exact physiological underpinnings of the measured FA changes remain to be elucidated. As expected, these changes are very small as compared to the physiological noise and a reliable detection of the signal change would require a large number of measurements. However, a substantial increase in signal-to-noise ratio was achieved by pooling the signal over the complete fiber tract. Adopting such a tract-based statistics enabled us to measure the signal within a practically feasible time period. Activation in the sensory thalamocortical tract and optic radiation in eight healthy human subjects was found during tactile and visual stimulation, respectively. Conclusions/Significance The results of our experiments indicate that these FA changes may serve as a functional contrast mechanism for white matter. This noninvasive fDTI method may provide a new approach to study functional neural networks in the human brain.
Collapse
Affiliation(s)
- René C W Mandl
- Rudolf Magnus Institute of Neuroscience, University Medical Center Utrecht, Utrecht, The Netherlands.
| | | | | | | | | | | |
Collapse
|
26
|
Chardonnet S, Le Marechal P, Cheval H, Le Caer JP, Decottignies P, Laprevote O, Laroche S, Davis S. Large-scale study of phosphoproteins involved in long-term potentiation in the rat dentate gyrusin vivo. Eur J Neurosci 2008; 27:2985-98. [DOI: 10.1111/j.1460-9568.2008.06280.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
27
|
Hemdan S, Almazan G. Dopamine-induced toxicity is synergistically potentiated by simultaneous HSP-90 and Akt inhibition in oligodendrocyte progenitors. J Neurochem 2008; 105:1223-34. [DOI: 10.1111/j.1471-4159.2008.05227.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
28
|
Broberg M, Pope KJ, Lewis T, Olsson T, Nilsson M, Willoughby JO. Cell swelling precedes seizures induced by inhibition of astrocytic metabolism. Epilepsy Res 2008; 80:132-41. [PMID: 18440781 DOI: 10.1016/j.eplepsyres.2008.03.012] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2007] [Revised: 03/12/2008] [Accepted: 03/13/2008] [Indexed: 10/22/2022]
Abstract
It is currently unknown what processes take place at the interface between non-ictal and ictal activity during seizure initiation. In this study, using paralysed awake rats, we focally inhibited astrocytic metabolism with fluorocitrate (FC), causing seizures. We measured changes in electroencephalogram (EEG) (0-300 Hz), and extracellular ion-concentrations during ictal onsets defining possible relationships with impedance-determined cell swelling. In animals showing ictal activity (69%) there were spike-wave discharges, spike-wave discharges followed by spreading depression and spreading depression without any discharges. In a high proportion of spike-wave discharges (>95%), just prior to the first spike-wave discharge, there was a decrease in the volume of the extracellular space. Following the initiation of cell swelling and prior to discharges, there were increases in high-frequency (150-300 Hz) EEG activity, increases in extracellular potassium- and decreases in extracellular calcium-concentrations. We suggest that EEG and ionic changes are not causative of cell swelling. Cell swelling due to metabolic failure in astrocytes at the injected site may release excitatory amino acids. At the same time, our results suggest ion homeostasis is not maintained and increased neuronal excitability and synchronisation occur. These could be the drivers changing normal brain activity into ictal activity.
Collapse
Affiliation(s)
- Marita Broberg
- Center for Neuroscience and Department of Medicine, Flinders University, Adelaide 5001, South Australia, Australia.
| | | | | | | | | | | |
Collapse
|
29
|
Canals S, Beyerlein M, Keller AL, Murayama Y, Logothetis NK. Magnetic resonance imaging of cortical connectivity in vivo. Neuroimage 2008; 40:458-472. [PMID: 18222710 DOI: 10.1016/j.neuroimage.2007.12.007] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2007] [Revised: 11/22/2007] [Accepted: 12/03/2007] [Indexed: 11/26/2022] Open
Abstract
Magnetic resonance imaging of neuronal connectivity in vivo opens up the possibility of performing longitudinal investigations on neuronal networks. This is one main reason for the attention that paramagnetic ion manganese (Mn2+) has attracted as a potential anterograde neuronal tracer for MRI experiments. However, the correct and possibly repeated use of this tracer--or of any tracer for that matter, including heavy metals--requires the development of an administration strategy that minimizes its toxic effects. Here we first investigated the conditions that maximize the tracing efficiency of Mn2+ and preserve viability and tissue architectonics in combined MRI and histology experiments in rats. We demonstrate that most common protocols for neuronal tract tracing using Mn2+ result in large neuronal and glial lesions. The toxicity of manganese is distinct during intracortical injections and blocks the transfer of the tracer. After optimizing the technique, we could show that extensive cortical connectivity maps can be generated, with no sign of neuronal damage. Importantly, preservation of tissue viability improves the efficiency of Mn2+ in tracing neuronal connections. We have successfully used this technique to trace corticofugal somatosensory and motor pathways in individual animals and describe a connectivity index (CnI) based on Mn2+ transport that quantitatively reveals cortical heterogeneities in interhemispheric communication. Finally, we have significantly improved the resolution of the technique by continuously infusing very low concentrations of Mn2+ into the target area using osmotic pumps coupled to chronically implanted brain cannulae. The specific, nontoxic and quantitative nature of the neuronal tracings described here indicates the value of this tracer for chronic studies of development and plasticity as well as for studies of brain pathology.
Collapse
Affiliation(s)
- S Canals
- Max Planck Institute for Biological Cybernetics, 72076 Tübingen, Germany.
| | - M Beyerlein
- Max Planck Institute for Biological Cybernetics, 72076 Tübingen, Germany
| | - A L Keller
- Max Planck Institute for Biological Cybernetics, 72076 Tübingen, Germany
| | - Y Murayama
- Max Planck Institute for Biological Cybernetics, 72076 Tübingen, Germany
| | - N K Logothetis
- Max Planck Institute for Biological Cybernetics, 72076 Tübingen, Germany; Imaging Science and Biomedical Engineering University of Manchester, Manchester, UK.
| |
Collapse
|
30
|
Guidolin D, Fuxe K, Neri G, Nussdorfer GG, Agnati LF. On the role of receptor–receptor interactions and volume transmission in learning and memory. ACTA ACUST UNITED AC 2007; 55:119-33. [PMID: 17408566 DOI: 10.1016/j.brainresrev.2007.02.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2006] [Revised: 02/13/2007] [Accepted: 02/14/2007] [Indexed: 11/16/2022]
Abstract
Learning and memory seem to be inherent to a biological neural network. To emerge, they need an extensive functional connectivity, enabling a large repertoire of possible responses to stimuli, and sensitivity of the connectivity to activity, allowing for the selection of adaptive responses. According to the classical view about the organization of the CNS, the connectivity issue is realized by the huge amount of synaptic contacts each neuron establishes, while the adaptation of the network to specific tasks is obtained by mechanisms of activity-dependent synaptic plasticity. The discovery of direct receptor-receptor interactions at the level of the plasma membrane and the existence in the brain of two main modes of communication, the wiring transmission (such as the synaptic transmission) and the volume transmission (based on the diffusion of signals in the extracellular space), provided a broader view of the functional organization of the CNS with potential important consequences on the understanding of learning and memory processes. Owing to receptor-receptor interactions clusters of receptors, the receptor mosaics (RM), can be formed at the plasma membrane where they can work as collective functional units. As a consequence, the connections between the cells become themselves networks (molecular networks) able to adapt their function according to the stimuli they receive. Learning, therefore, can occur also at the level of RMs. Thus, memory formation seems not only to be a distributed process, but also to follow a hierarchical morpho-functional organization. Furthermore, the combination of the two different forms of transmission could allow processes of correlation and coordination to be established between networks and network elements without the need of additional physical connections, leading to a significant increase of the degrees of freedom available to the CNS for learning.
Collapse
Affiliation(s)
- Diego Guidolin
- Department of Human Anatomy and Physiology, Section of Anatomy, University of Padova Medical School, via Gabelli 65, I-35121 Padua, Italy.
| | | | | | | | | |
Collapse
|
31
|
Czéh B, Lucassen PJ. What causes the hippocampal volume decrease in depression? Are neurogenesis, glial changes and apoptosis implicated? Eur Arch Psychiatry Clin Neurosci 2007; 257:250-60. [PMID: 17401728 DOI: 10.1007/s00406-007-0728-0] [Citation(s) in RCA: 305] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Even though in vivo imaging studies document significant reductions of hippocampal volume in depressed patients, the exact underlying cellular mechanisms are unclear. Since stressful life events are associated with an increased risk of developing depression, preclinical studies in which animals are exposed to chronic stress have been used to understand the hippocampal shrinkage in depressed patients. Based on morphometrical studies in these models, parameters like dendritic retraction, suppressed adult neurogenesis and neuronal death, all due to elevated levels of glucocorticoids, have been suggested as major causative factors in hippocampal shrinkage. However, histopathological studies examining hippocampi of depressed individuals have so far failed to confirm either a massive neuronal loss or a suppression of dentate neurogenesis, an event that is notably very rare in adult or elderly humans. In fact, many of the structural changes and the volume reduction appear to be reversible. Clearly, more histopathological studies are needed; especially ones that (a) employ stereological quantification, (b) focus on specific cellular elements and populations, and (c) are performed in nonmedicated depressed patients. We conclude that mainly other factors, like alterations in the somatodendritic, axonal, and synaptic components and putative glial changes are most likely to explain the hippocampal shrinkage in depression, while shifts in fluid balance or changes in the extracellular space cannot be excluded either.
Collapse
Affiliation(s)
- Boldizsár Czéh
- Clinical Neurobiology Laboratory, German Primate Center, Kellnerweg 4, 37077 Göttingen, Germany.
| | | |
Collapse
|
32
|
Chvátal A, Anderová M, Kirchhoff F. Three-dimensional confocal morphometry - a new approach for studying dynamic changes in cell morphology in brain slices. J Anat 2007; 210:671-83. [PMID: 17488344 PMCID: PMC2375758 DOI: 10.1111/j.1469-7580.2007.00724.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Pathological states in the central nervous system lead to dramatic changes in the activity of neuroactive substances in the extracellular space, to changes in ionic homeostasis and often to cell swelling. To quantify changes in cell morphology over a certain period of time, we employed a new technique, three-dimensional confocal morphometry. In our experiments, performed on enhanced green fluorescent protein/glial fibrillary acidic protein astrocytes in brain slices in situ and thus preserving the extracellular microenvironment, confocal morphometry revealed that the application of hypotonic solution evoked two types of volume change. In one population of astrocytes, hypotonic stress evoked small cell volume changes followed by a regulatory volume decrease, while in the second population volume changes were significantly larger without subsequent volume regulation. Three-dimensional cell reconstruction revealed that even though the total astrocyte volume increased during hypotonic stress, the morphological changes in various cell compartments and processes were more complex than have been previously shown, including swelling, shrinking and structural rearrangement. Our data show that astrocytes in brain slices in situ during hypotonic stress display complex behaviour. One population of astrocytes is highly capable of cell volume regulation, while the second population is characterized by prominent cell swelling, accompanied by plastic changes in morphology. It is possible to speculate that these two astrocyte populations play different roles during physiological and pathological states.
Collapse
Affiliation(s)
- Alexandr Chvátal
- Department of Cellular Neurophysiology, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Prague, Czech Republic.
| | | | | |
Collapse
|
33
|
Chvátal A, Anderová M, Hock M, Prajerová I, Neprasová H, Chvátal V, Kirchhoff F, Syková E. Three-dimensional confocal morphometry reveals structural changes in astrocyte morphology in situ. J Neurosci Res 2007; 85:260-71. [PMID: 17086549 DOI: 10.1002/jnr.21113] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Neuronal activity and many pathological states in the CNS are accompanied by transient astrocytic swelling, which affects excitability, extrasynaptic transmission, and neuron-glia interactions. By using three-dimensional confocal morphometry (3DCM), we quantified the morphometric parameters of astrocytes in intact tissue. In experiments performed in brain cortex slices from transgenic GFAP/EGFP mice, we applied 3DCM to study the dynamic changes in astrocyte morphology during hypotonic stress. Our morphometric analysis showed that the effect of a 10-min application of hypotonic solution (200 mmol/kg) on the swelling of different cell compartments was dependent on the extent of the swelling of the total astrocyte volume. If the swelling of the whole cell, i.e., soma and processes, was less than approximately 10%, there were no differences between the swelling of the soma and the processes. However, if the swelling of the total cell volume was greater than 10%, the swelling of the processes was greater than the swelling of the soma. Analyzing the effect of hypotonic solution on the morphology of these astrocytes revealed that the total cell volume increased; however, certain cell compartments were distinguished in which the volume increased, whereas in other compartments cell volume decreased or apparently did not change, and the structure of some compartments was altered. Our data show that astrocytes in brain slices undergoing hypotonic stress display cell volume regulation as well as transient changes in morphology.
Collapse
Affiliation(s)
- Alexandr Chvátal
- Department of Neurobiology, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Prague, Czech Republic.
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Hemdan S, Almazan G. Deficient peroxide detoxification underlies the susceptibility of oligodendrocyte progenitors to dopamine toxicity. Neuropharmacology 2007; 52:1385-95. [PMID: 17400258 DOI: 10.1016/j.neuropharm.2007.01.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2006] [Revised: 01/24/2007] [Accepted: 01/30/2007] [Indexed: 10/23/2022]
Abstract
Oligodendrocyte progenitors are highly susceptible to oxidative stress due to their limited content of antioxidants and high iron levels. We previously showed that iron plays a central role in the toxicity of dopamine (DA) to oligodendrocyte progenitors. Here, we further explore the mechanisms involved in DA toxicity, specifically the role of superoxide and the glutathione system. DA induces accumulation of superoxide, membrane damage and loss in cell viability. An iron chelator, deferoxamine, reduces superoxide accumulation. However, a superoxide dismutase mimetic, MnTBAP, potentiates DA toxicity, suggesting that superoxide plays an indirect role in toxicity through dismutation to H2O2. In addition, the glutathione (GSH) analog (GME), blocks DA-induced superoxide accumulation, heme-oxygenase-1 (HO-1) expression and caspase-3 activation, and reduces cell death, while the glutathione synthetase inhibitor, buthionine sulfoximine, potentiates DA-induced HO-1 expression and cell death. Moreover, a mimetic of the peroxide-scavenging enzyme, glutathione peroxidase (GPx), ebselen, blocks caspase-3 activation induced by DA alone or in combination with iron. In conclusion, superoxide and inadequate defense by glutathione and GPx are responsible for the susceptibility of oligodendrocyte progenitors to DA toxicity. Furthermore, peroxides play a primary role in toxicity induced by DA and iron.
Collapse
Affiliation(s)
- Sandy Hemdan
- Department of Pharmacology and Therapeutics, McGill University, 3655 Promenade Sir William Osler, Room 1321, Montreal, QC H3G 1Y6, Canada
| | | |
Collapse
|
35
|
Mulligan SJ, MacVicar BA. VRACs CARVe a path for novel mechanisms of communication in the CNS. ACTA ACUST UNITED AC 2006; 2006:pe42. [PMID: 17047222 DOI: 10.1126/stke.3572006pe42] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Because the brain is encased by the skull, the ability to control cell volume in the brain is crucial, and pathological conditions that disturb cell volume homeostasis may severely compromise neural function and survival. Astrocytes are the main cell type to show swelling in response to pathological conditions. More recently, a role for swelling-induced neurotransmitter release from astrocytes under nonpathological conditions has been reported. Astrocytes express a volume-regulated anion channel (VRAC) that is involved in volume homeostasis. In addition to transporting chloride, VRACs allow the efflux of chloride and amino acids such as taurine, glutamate, and aspartate. Glutamate and aspartate are potent activators of neuronal glutamate receptors. Therefore, this nonsynaptic form of cellular communication may modulate neuronal excitability and synaptic activity.
Collapse
Affiliation(s)
- Sean J Mulligan
- Department of Physiology, University of Saskatchewan, Saskatoon, SK, Canada S7N 5E5
| | | |
Collapse
|
36
|
Kilb W, Dierkes PW, Syková E, Vargová L, Luhmann HJ. Hypoosmolar conditions reduce extracellular volume fraction and enhance epileptiform activity in the CA3 region of the immature rat hippocampus. J Neurosci Res 2006; 84:119-29. [PMID: 16634058 DOI: 10.1002/jnr.20871] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The osmolarity of the extracellular space (ECS) compartment is an important factor determining the excitability of neuronal tissue. In the adult hippocampus an important role of osmolarity and ECS diffusion parameters on the susceptibility to epileptic events is well established, but the influence of hypo- and hyperosmolar conditions on the immature hippocampus remains elusive. To investigate the influence of osmolarity on epileptiform activity, extracellular field potentials were recorded in the CA3 region of hippocampal slices of immature (postnatal days 4-7) Wistar rats. The ECS diffusion parameters were determined by the real-time tetramethylammonium (TMA+) iontophoretic method with ion-selective microelectrodes in immature hippocampal slices and showed a lack of diffusion anisotropy; a tortuosity of about 1.39; and a volume fraction, alpha, of 0.41 +/- 0.01 (n = 10 slices). A reduction in osmolarity of -90 mOsm induced a decrease in alpha to 0.17 +/- 0.02 (n = 4 slices). The frequency of epileptiform activity elicited in 10-50 microM 4-AP-containing low-Mg2+ solution was increased under -90 mOsm and -40 mOsm hypoosmolar conditions by 39.9% +/- 8.1% (n = 16) and 24.1% +/- 4.8% (n = 10), respectively, whereas hyperosmolar solutions decreased the frequency. A -90-mOsm reduction in the osmolarity of low-Mg2+ solution induced epileptiform activity in nine of 19 slices. In summary, these results demonstrate that hypoosmolar conditions increased excitability and susceptibility to epileptiform activity in immature hippocampal slices, suggesting a functional role of the larger alpha in suppression of seizures.
Collapse
Affiliation(s)
- Werner Kilb
- Institute of Physiology and Pathophysiology, Johannes Gutenberg-University, Mainz, Germany.
| | | | | | | | | |
Collapse
|
37
|
Hemdan S, Almazan G. Iron contributes to dopamine-induced toxicity in oligodendrocyte progenitors. Neuropathol Appl Neurobiol 2006; 32:428-40. [PMID: 16866988 DOI: 10.1111/j.1365-2990.2006.00757.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Iron is potentially toxic to oligodendrocyte progenitors due to its high intracellular levels and its ability to catalyse oxidant-producing reactions. Oxidative stress resulting from a hypoxic-ischaemic insult has been implicated in death of oligodendrocyte progenitors that occurs in the hypomyelinating disorder periventricular leucomalacia. Ischaemic insults induce the release of various neurotransmitters, including dopamine (DA), and we previously showed that DA is toxic to cultured oligodendrocytes, by inducing oxidative stress and apoptosis. Therefore, we investigated the role of iron in DA-induced cell death in oligodendrocyte progenitors. Intracellular iron levels were altered using an iron chelator, deferoxamine (DFO), and supplementation with ferrous sulphate (FeSO(4)). Addition of FeSO(4) to cultures increased DA-induced toxicity as assessed by mitochondrial dehydrogenase activity and cellular release of lactate dehydrogenase. Furthermore, FeSO(4) increased expression of the stress protein heme oxygenase-1 (HO-1), nuclear condensation and caspase-3 activation. In contrast, preincubation with DFO reduced these events as well as cleavage of alpha-spectrin, a caspase-3 substrate. In addition, FeSO(4) reversed the protective effect of DFO on DA-induced cytotoxicity, HO-1 expression and caspase-3 activation. These results indicate that elevated levels of free iron contribute to DA-induced toxicity in oligodendrocyte progenitors.
Collapse
Affiliation(s)
- S Hemdan
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | | |
Collapse
|
38
|
Abstract
Neuronal-glial circuits underlie integrative processes in the nervous system. Function of glial syncytium is, to a very large extent, regulated by the intracellular calcium signaling system. Glial calcium signals are triggered by activation of multiple receptors, expressed in glial membrane, which regulate both Ca2+ entry and Ca2+ release from the endoplasmic reticulum. The endoplasmic reticulum also endows glial cells with intracellular excitable media, which is able to produce and maintain long-ranging signaling in a form of propagating Ca2+ waves. In pathological conditions, calcium signals regulate glial response to injury, which might have both protective and detrimental effects on the nervous tissue.
Collapse
|
39
|
Homola A, Zoremba N, Slais K, Kuhlen R, Syková E. Changes in diffusion parameters, energy-related metabolites and glutamate in the rat cortex after transient hypoxia/ischemia. Neurosci Lett 2006; 404:137-42. [PMID: 16759801 DOI: 10.1016/j.neulet.2006.05.028] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2006] [Revised: 05/12/2006] [Accepted: 05/14/2006] [Indexed: 11/29/2022]
Abstract
It has been shown that global anoxia leads to dramatic changes in the diffusion properties of the extracellular space (ECS). In this study, we investigated how changes in ECS volume and geometry in the rat somatosensory cortex during and after transient hypoxia/ischemia correlate with extracellular concentrations of energy-related metabolites and glutamate. Adult male Wistar rats (n = 12) were anesthetized and subjected to hypoxia/ischemia for 30 min (ventilation with 10% oxygen and unilateral carotid artery occlusion). The ECS diffusion parameters, volume fraction and tortuosity, were determined from concentration-time profiles of tetramethylammonium applied by iontophoresis. Concentrations of lactate, glucose, pyruvate and glutamate in the extracellular fluid (ECF) were monitored by microdialysis (n = 9). During hypoxia/ischemia, the ECS volume fraction decreased from initial values of 0.19 +/- 0.03 (mean +/- S.E.M.) to 0.07 +/- 0.01 and tortuosity increased from 1.57 +/- 0.01 to 1.88 +/- 0.03. During reperfusion the volume fraction returned to control values within 20 min and then increased to 0.23 +/- 0.01, while tortuosity only returned to original values (1.53 +/- 0.06). The concentrations of lactate and glutamate, and the lactate/pyruvate ratio, substantially increased during hypoxia/ischemia, followed by continuous recovery during reperfusion. The glucose concentration decreased rapidly during hypoxia/ischemia with a subsequent return to control values within 20 min of reperfusion. We conclude that transient hypoxia/ischemia causes similar changes in ECS diffusion parameters as does global anoxia and that the time course of the reduction in ECS volume fraction correlates with the increase of extracellular concentration of glutamate. The decrease in the ECS volume fraction can therefore contribute to an increased accumulation of toxic metabolites, which may aggravate functional deficits and lead to damage of the central nervous system (CNS).
Collapse
Affiliation(s)
- Ales Homola
- Department of Neuroscience and Centre for Cell Therapy and Tissue Repair, 2nd Medical Faculty, Prague, Czech Republic
| | | | | | | | | |
Collapse
|
40
|
Cui QL, Fogle E, Almazan G. Muscarinic acetylcholine receptors mediate oligodendrocyte progenitor survival through Src-like tyrosine kinases and PI3K/Akt pathways. Neurochem Int 2006; 48:383-93. [PMID: 16439036 DOI: 10.1016/j.neuint.2005.11.014] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2005] [Revised: 11/22/2005] [Accepted: 11/29/2005] [Indexed: 01/22/2023]
Abstract
The function of muscarinic acetylcholine receptors expressed in oligodendrocytes and in myelin has remained largely undetermined. Here we present evidence that incubation of oligodendrocyte progenitors, deprived of growth factor, with the acetylcholine analog carbachol significantly reduced cell death by apoptosis and blocked caspase-3 cleavage. This protective effect was reversed by atropine, a muscarinic acetylcholine receptor antagonist, as well as by specific inhibitors of intracellular signaling molecules, including phosphatidylinositol 3-kinase (Wortmannin and LY294002), Akt (Akt inhibitor III) and Src-like tyrosine kinases (PP2), but not by the mitogen-activated protein kinase kinase inhibitor, PD98059. Activation of Akt by carbachol was antagonized by atropine and inhibited by LY294002 and PP2. The Src-like tyrosine kinase inhibitor, PP2, also reduced carbachol stimulation of extracellular signal-regulated kinases 1/2 and cAMP-response element binding protein in a dose-dependent manner. Furthermore, carbachol increased tyrosine-phosphorylation of Fyn, a member of the Src-like tyrosine kinases. These results indicate that muscarinic acetylcholine receptors play an important role in oligodendrocyte progenitor survival through transduction pathways involving activation of Src-like tyrosine kinases and phosphatidylinositol 3-kinase/Akt.
Collapse
Affiliation(s)
- Qiao-Ling Cui
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Que., Canada
| | | | | |
Collapse
|
41
|
Fan Y, Zhang J, Sun XL, Gao L, Zeng XN, Ding JH, Cao C, Niu L, Hu G. Sex- and region-specific alterations of basal amino acid and monoamine metabolism in the brain of aquaporin-4 knockout mice. J Neurosci Res 2006; 82:458-64. [PMID: 16237719 DOI: 10.1002/jnr.20664] [Citation(s) in RCA: 96] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Aquaporin-4 (AQP4), a predominant water channel of the brain, mediates transmembrane water movement at the blood-brain barrier and brain-cerebrospinal fluid interface. A broad pattern of evidence indicates that AQP4 and regulators of its expression are potential targets for treatment of brain swelling, but whether it participates in the regulation of neurotransmission has not been reported. We examined neurochemical differences between AQP4-knockout and wild-type mice with particular focus on neurotransmission. Basal tissue neurotransmitter and metabolite levels were measured by high-performance liquid chromatography. Significant sex- and region-specific differences of amino acids and monoamines were found in the brain of wild-type and AQP4-knockout mice. In cortex, striatum, and hippocampus of male AQP4-knockout mice, an increase of glutamine and decrease of aspartate were observed. Glutamate was increased only in female AQP4-knockout mice. The lack of AQP4 failed to affect the levels of gamma-aminobutyric acid and taurine. In the medial prefrontal cortex of AQP4-knockout mice, the levels of serotonin and norepinephrine were increased, but no significant change in dopamine level was found. In the striatum of male AQP4-knockout mice, the levels of dopamine and serotonin were remarkably increased, which was not found in female mice. In the hypothalamus of AQP4-knockout mice, only the serotonin level was altered. These results provide the first evidence that the lack of AQP4 expression is accompanied by sex- and region-specific alterations in brain amino acid and monoamine metabolism.
Collapse
Affiliation(s)
- Yi Fan
- Laboratory of Neuropharmacology, Department of Anatomy, Histology and Pharmacology, Institute of Neurosciences, Nanjing Medical University, Jiangsu, China
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Abstract
Recent discoveries suggest that astrocytes are an integral part of synaptic connections, as they sense and modulate synaptic activity. Moreover, there is evidence that astrocytes change the number of synaptic connections directly via synaptogenic signals or indirectly, by modifying the morphology of axons and dendrites. Here, we formulate the hypothesis that astrocytes mediate the morphological homeostasis of nerve cells, which is any adaptation of the morphology of a neuron to preserve its ability to respond to and generate synaptic activity during learning and memory-induced changes. We argue that astrocytes control neuronal morphology locally and across long-ranging assemblies of neurons and that on the other hand, astrocytes are part of the engram with plasticity-related changes affecting their morphology.
Collapse
Affiliation(s)
- Michal Slezak
- Max-Planck/CNRS Group, UPR 2356, Centre de Neurochimie, 5, rue Blaise Pascal, 67084 Strasbourg Cedex, France
| | | | | |
Collapse
|
43
|
Syková E. Extrasynaptic volume transmission and diffusion parameters of the extracellular space. Neuroscience 2005; 129:861-76. [PMID: 15561404 DOI: 10.1016/j.neuroscience.2004.06.077] [Citation(s) in RCA: 181] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/30/2004] [Indexed: 10/26/2022]
Abstract
Extrasynaptic communication between neurons or neurons and glia is mediated by the diffusion of neuroactive substances in the volume of the extracellular space (ECS). The size and irregular geometry of the diffusion channels in the ECS substantially differ not only around individual cells but also in different CNS regions and thus affect and direct the movement of various neuroactive substances in the ECS. Diffusion in the CNS is therefore not only inhomogeneous, but often also anisotropic. The diffusion parameters in adult mammals (including humans), ECS volume fraction alpha (alpha=ECS volume/total tissue volume) and tortuosity lambda (lambda(2)=free/apparent diffusion coefficient), are typically 0.20-0.25 and 1.5-1.6, respectively, and as such hinder the diffusion of neuroactive substances and water. These diffusion parameters modulate neuronal signaling, neuron-glia communication and extrasynaptic "volume" transmission. A significant decrease in ECS volume fraction and an increase in diffusion barriers (tortuosity) occur during neuronal activity and pathological states. The changes are often related to cell swelling, cell loss, astrogliosis, the rearrangement of neuronal and astrocytic processes and changes in the extracellular matrix. They are also altered during physiological states such as development, lactation and aging. Plastic changes in ECS volume, tortuosity and anisotropy significantly affect neuron-glia communication, the spatial relation of glial processes toward synapses, glutamate or GABA "spillover" and synaptic crosstalk. The various changes in tissue diffusivity occurring during many pathological states are important for diagnosis, drug delivery and treatment.
Collapse
Affiliation(s)
- E Syková
- Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Prague and Department of Neuroscience, Charles University, Second Medical Faculty, Vídenská 1083, 14220 Prague 4, Czech Republic.
| |
Collapse
|
44
|
Nagelhus EA, Mathiisen TM, Ottersen OP. Aquaporin-4 in the central nervous system: cellular and subcellular distribution and coexpression with KIR4.1. Neuroscience 2005; 129:905-13. [PMID: 15561407 DOI: 10.1016/j.neuroscience.2004.08.053] [Citation(s) in RCA: 380] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/23/2004] [Indexed: 12/16/2022]
Abstract
Aquaporin-4 (AQP4) is the predominant water channel in the neuropil of the central nervous system. It is expressed primarily in astrocytes, but also occurs in ependymocytes and endothelial cells. A striking feature of AQP4 expression is its polarized distribution in brain astrocytes and retinal Muller cells. Thus, immunogold analyses have revealed an enrichment of AQP4 in endfeet membranes in contact with brain microvessels or subarachnoidal space and a low but significant concentration in non-endfeet membranes, including those astrocyte membranes that ensheath glutamate synapses. The subcellular compartmentation of AQP4 mimics that of the potassium channel Kir4.1, which is implicated in spatial buffering of K(+). We propose that AQP4 works in concert with Kir4.1 and the electrogenic bicarbonate transporter NBC and that water flux through AQP4 contributes to the activity dependent volume changes of the extracellular space. Such volume changes are important as they affect the extracellular solute concentrations and electrical fields, and hence neuronal excitability. We conclude that AQP4-mediated water flux represents an integral element of brain volume and ion homeostasis.
Collapse
Affiliation(s)
- E A Nagelhus
- Nordic Centre of Excellence for Research in Water Imbalance Related Disorders and Centre for Molecular Biology and Neuroscience, Institute of Basic Medical Sciences, University of Oslo, POB 1105 Blindern, N-0317 Oslo, Norway.
| | | | | |
Collapse
|
45
|
Anderová M, Antonova T, Petrík D, Neprasová H, Chvátal A, Syková E. Voltage-dependent potassium currents in hypertrophied rat astrocytes after a cortical stab wound. Glia 2005; 48:311-26. [PMID: 15390116 DOI: 10.1002/glia.20076] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Changes in the membrane properties of reactive astrocytes in gliotic cortex induced by a stab wound were studied in brain slices of 21-28-day-old rats, using the patch-clamp technique and were correlated with changes in resting extracellular K+ concentration ([K+]e) measured in vivo using K+-selective microelectrodes. Based on K+ current expression, three types of astrocytes were identified in gliotic cortex: A1 astrocytes expressing a time- and voltage-independent K+ current component and additional inwardly rectifying K+ currents (K(IR)); A2 astrocytes expressing a time- and voltage-independent K+ current component and additional delayed outwardly rectifying K+ currents (K(DR)); and complex astrocytes expressing K(DR), K(IR), and A-type K+ (K(A)) currents and Na+ currents (I(Na)). Nestin/bromodeoxyuridine (BrdU)-negative A1 astrocytes were found further than approximately 100 microm from the stab wound and showed an upregulation of K(IR) currents within the first day post-injury (PI), correlating with an increased resting [K+]e. Their number declined from 62% of total astrocytes in control rats to 41% in rats at 7 days PI. Nestin/BrdU-positive A2 astrocytes were found only within a distance of approximately 100 microm from the stab wound and, in comparison to those in control rats, showed an upregulation of K(DR) currents. Their number increased from 8% of the total number of astrocytes in control rats to 39% 7 days PI. Both A1 and A2 astrocytes showed hypertrophied processes and increased GFAP staining, but an examination of cell morphology revealed greater changes in the surface/volume ratio in A2 astrocytes than in A1 astrocytes. Complex astrocytes did not display a hypertophied morphology; K(IR) currents in these cells were upregulated within 1 day PI, while the K(DR), K(A), and I(Na) currents were increased only 6 h PI. We conclude that two electrophysiologically, immunohistochemically, and morphologically distinct types of hypertrophied astrocytes are present at the site of a stab wound, depending on the distance from the lesion, and may have different functions in ionic homeostasis and/or regeneration.
Collapse
Affiliation(s)
- Miroslava Anderová
- Department of Neuroscience, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Prague, Czech Republic.
| | | | | | | | | | | |
Collapse
|
46
|
Kimelberg HK. Astrocytic swelling in cerebral ischemia as a possible cause of injury and target for therapy. Glia 2005; 50:389-397. [PMID: 15846797 DOI: 10.1002/glia.20174] [Citation(s) in RCA: 202] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
In this viewpoint article, I summarize data showing that the astrocytic swelling that occurs early after the acute CNS pathologies ischemia and traumatic brain injury is damaging. We have proposed that one reason may be the release of excitatory amino acids (EAA) via volume-activated anion channels (VRACs) that are activated by such swelling. This release could be a target for therapy, which could involve blocking the astrocytic swelling or the release mechanisms. The transport mechanisms likely causing the early astrocytic swelling are therefore summarized. In terms of targeting the release mechanisms, we have found a potent inhibitor of VRACs, tamoxifen, to be strongly neuroprotective in focal ischemia with a therapeutic window of 3 h after initiation of the ischemia. The question, however, of whether neuroprotection by tamoxifen can be solely attributed to VRAC inhibition in astrocytes has yet to be resolved.
Collapse
Affiliation(s)
- Harold K Kimelberg
- Neural and Vascular Biology, Ordway Research Institute, Albany, New York
| |
Collapse
|
47
|
Fuchs E, Czéh B, Kole MHP, Michaelis T, Lucassen PJ. Alterations of neuroplasticity in depression: the hippocampus and beyond. Eur Neuropsychopharmacol 2004; 14 Suppl 5:S481-90. [PMID: 15550346 DOI: 10.1016/j.euroneuro.2004.09.002] [Citation(s) in RCA: 169] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Early hypotheses on the pathophysiology of major depression were based on aberrant intrasynaptic concentrations of mainly the neurotransmitters serotonin and norepinephrine. However, recent neuroimaging studies have demonstrated selective structural changes across various limbic and nonlimbic circuits in the brains of depressed patients. In addition, postmortem morphometric studies revealed decreased glial and neuron densities in selected brain structures supporting the idea that major depression may be related to impairments of structural plasticity. Stressful life events are among the major predisposing risk factors for developing depression. Using the chronic psychosocial stress paradigm in male tree shrews, an animal model with a high validity for the pathophysiology of depressive disorders, we found that 1 month of stress reduced the in vivo concentrations of the brain metabolites N-acetyl-aspartate, choline-containing compounds, and (phospho)-creatine, as well as the proliferation rate in the dentate gyrus and the hippocampal volume. Even though long-lasting social conflict does not lead to a loss of principal cells, the hippocampal changes were accompanied by modifications in the incidence of apoptosis. Notably, these suppressive effects of social conflict on hippocampal structure could be counteracted by treatment with the antidepressant tianeptine. These findings support current theories proposing that major depressive disorders may be associated with impairment of structural plasticity and neural cellular resilience, and that antidepressants may act by correcting this dysfunction.
Collapse
Affiliation(s)
- Eberhard Fuchs
- Clinical Neurobiology Laboratory, German Primate Center, Kellnerweg 4, 37077 Göttingen, Germany.
| | | | | | | | | |
Collapse
|
48
|
Adermark L, Olsson T, Hansson E. Ethanol acutely decreases astroglial gap junction permeability in primary cultures from defined brain regions. Neurochem Int 2004; 45:971-8. [PMID: 15337295 DOI: 10.1016/j.neuint.2004.06.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2004] [Revised: 03/15/2004] [Accepted: 06/21/2004] [Indexed: 11/21/2022]
Abstract
The acute effect of hyperosmotic ethanol on gap junction permeability was examined in astroglial cells in primary culture from five different brain regions. Gap junction permeability was analyzed by measuring dye spreading from cell to cell with the low molecular weight dye Lucifer Yellow. Ethanol concentrations 25-300 mM significantly decreased dye spreading in cultures from the cerebral cortex in a dose-dependent but time-independent manner for up to 60 min. Besides cerebral cortex, exposure to 150 mM ethanol decreased dye spreading in astroglial cultures from the hippocampus and from the brain stem, while cultures from the olfactory bulb and from the hypothalamus were not significantly affected. The ethanol-induced decrease in dye spreading in cultures from the cerebral cortex was not mediated through changes in cell volume, osmolarity, protein kinase C (PKC) phosphorylation, intracellular pH, or intracellular calcium concentration ([Ca(2+)](i)). The decrease in dye spreading was abolished upon incubation in sodium-reduced buffer, and after blockage of the Na(+)/K(+)/2Cl(-) cotransporter with furosemide. The results presented here indicate that ethanol-mediated decrease in dye spreading is directly or indirectly dependent on sodium.
Collapse
Affiliation(s)
- Louise Adermark
- Institute of Clinical Neuroscience, Göteborg University, Box 420, SE 405 30 Göteborg, Sweden.
| | | | | |
Collapse
|
49
|
Abstract
Extrasynaptic transmission between neurons and communication between neurons and glia are mediated by the diffusion of neuroactive substances in the extracellular space (ECS)--volume transmission. Diffusion in the CNS is inhomogeneous and often not uniform in all directions (anisotropic). Ionic changes and amino acid release result in cellular (particularly glial) swelling, compensated for by ECS shrinkage and a decrease in the apparent diffusion coefficients of neuroactive substances or water (ADCW). The diffusion parameters of the CNS in adult mammals (including humans), ECS volume fraction alpha (alpha = ECS volume/total tissue volume; normally 0.20-0.25) and tortuosity lambda (lambda2 = D/ADC; normally 1.5-1.6), hinder the diffusion of neuroactive substances and water. A significant decrease in ECS volume and an increase in diffusion barriers (tortuosity) and anisoptropy have been observed during stimulation, lactation or learning deficits during aging, due to structural changes such as astrogliosis, the re-arrangement of astrocytic processes and a loss of extracellular matrix. Decreases in the apparent diffusion coefficient of tetramethylammonium (ADCTMA) and ADCW due to astrogliosis and increased proteoglycan expression were found in the brain after injury and in grafts of fetal tissue. Tenascin-R and tenascin C-deficient mice also showed significant changes in ADCTMA and ADCW, suggesting an important role for extracellular matrix molecules in ECS diffusion. Changes in ECS volume, tortuosity and anisotropy significantly affect neuron-glia communication, the spatial relation of glial processes towards synapses, the efficacy of glutamate or GABA 'spillover' and synaptic crosstalk, the migration of cells, the action of hormones and the toxic effects of neuroactive substances and can be important for diagnosis, drug delivery and new treatment strategies.
Collapse
Affiliation(s)
- Eva Syková
- Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Videnska 1083, 14220 Prague 4, Czech Republic.
| |
Collapse
|
50
|
Ryabov SI. Changes in the volume of the intercellular space of the cerebral cortex in conditions of peripheral stimulation in rats. ACTA ACUST UNITED AC 2004; 34:491-3. [PMID: 15330288 DOI: 10.1023/b:neab.0000022635.03250.95] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Changes in the volume of the intercellular space of the rat cerebral cortex in response to peripheral repetitive stimulation were studied. The volume of the intercellular space and its changes were assessed by a modification of the four-electrode impedance method. The results suggest that evoked electrical activity in the cerebral cortex was accompanied by 3-5% decreases in the volume of the intercellular space.
Collapse
Affiliation(s)
- S I Ryabov
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, 5a Butlerov Street, 117865 Moscow, Russia
| |
Collapse
|