3601
|
Jehs T, Faber C, Juel HB, Nissen MH. Astrocytoma cells upregulate expression of pro-inflammatory cytokines after co-culture with activated peripheral blood mononuclear cells. APMIS 2011; 119:551-61. [PMID: 21749456 DOI: 10.1111/j.1600-0463.2011.02773.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
In this study, we investigated the effect of CD3/CD28-activated peripheral blood mononuclear cells (PBMCs) on two human astrocytoma cell lines (1321N1 and CCF-STTG1), with respect to the expression of cytokines and chemokines. We used an in vitro co-culture system in which the astrocytoma cells and PBMCs were separated by a membrane in a transwell system. Proliferation of T cells was quantified by a [(3) H]-thymidine incorporation assay. Differential gene expression of astrocytoma cells cultured alone or with PBMCs was identified using gene arrays. Protein expression of CCL3, CCL5, CXCL8, CXCL9, CXCL10, IL6, and IL1β was determined by Luminex. Co-culture of activated T cells and astrocytoma cells resulted in inhibition of T cell proliferation. Moreover, astrocytoma cells upregulated a number of pro-inflammatory genes including CCL3, CCL5, CXCL8, CXCL9, CXCL10, IL6, and IL1β upon co-culture with activated PBMCs. These results were confirmed on the protein level. Astrocytoma cells inhibited the proliferation of T cells, indicating that astrocytes may play an important role in controlling T cell mediated immune responses in the brain. Contrary to this, soluble factors derived from CD3/CD28-activated PBMCs affected the transcriptome of astrocytoma cells, which then upregulated several pro-inflammatory genes and proteins. This could be part of neuroinflammatory events and may play a role in neurodegenerative diseases.
Collapse
Affiliation(s)
- Tina Jehs
- Eye Research Unit, Department of International Health, Immunology and Microbiology, University of Copenhagen, Denmark.
| | | | | | | |
Collapse
|
3602
|
Barreto GE, Gonzalez J, Torres Y, Morales L. Astrocytic-neuronal crosstalk: implications for neuroprotection from brain injury. Neurosci Res 2011; 71:107-13. [PMID: 21693140 DOI: 10.1016/j.neures.2011.06.004] [Citation(s) in RCA: 154] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2011] [Revised: 04/28/2011] [Accepted: 05/16/2011] [Indexed: 10/18/2022]
Abstract
The older neurocentric view of the central nervous system (CNS) has changed radically with the growing understanding of the many essential functions of astrocytes. Advances in our understanding of astrocytes include new observations about their structure, organization, function and supportive actions to other cells. Although the contribution of astrocytes to the process of brain injury has not been clearly defined, it is thought that their ability to provide support to neurons after cerebral damage is critical. Astrocytes play a fundamental role in the pathogenesis of brain injury-associated neuronal death, and this secondary injury is primarily a consequence of the failure of astrocytes to support the essential metabolic needs of neurons. These needs include K+ buffering, glutamate clearance, brain antioxidant defense, close metabolic coupling with neurons, and the modulation of neuronal excitability. In this review, we will focus on astrocytic activities that can both protect and endanger neurons, and discuss how manipulating these functions provides a novel and important strategy to enhance neuronal survival and improve the outcome following brain injury.
Collapse
Affiliation(s)
- George E Barreto
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá DC, Colombia.
| | | | | | | |
Collapse
|
3603
|
Blockade of P2 nucleotide receptors after spinal cord injury reduced the gliotic response and spared tissue. J Mol Neurosci 2011; 46:167-76. [PMID: 21647706 DOI: 10.1007/s12031-011-9567-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2011] [Accepted: 05/23/2011] [Indexed: 12/22/2022]
Abstract
Spinal cord injury (SCI) triggers a sequel of events commonly associated with cell death and dysfunction of glias and neurons surrounding the lesion. Although astrogliosis and glial scar formation have been involved in both damage and repair processes after SCI, their role remains controversial. Our goal was to investigate the effects of the P2 receptors antagonists, PPADS and suramin, in the establishment of the reactive gliosis and the formation of the glial scar. Molecular biology, immunohistochemistry, spared tissue, and locomotor behavioral studies were used to evaluate astrogliosis, in adult female Sprague-Dawley rats treated with P2 antagonists after moderate injury with the NYU impactor device. Semi-quantitative RT-PCR confirmed the presence of P2Y(1,) P2Y(2,) P2Y(4,) P2Y(6,) P2Y(12), and P2X(2) receptors in the adult spinal cord. Immunohistochemistry studies confirmed a significant decrease in GFAP-labeled cells at the injury epicenter as well as a decrease in spared tissue after treatment with the antagonists. Functional open field testing revealed no significant locomotor score differences between treated and control animals. Our work is consistent with studies suggesting that astrogliosis is an important event after SCI that limits tissue damage and lesion spreading.
Collapse
|
3604
|
Halliday GM, Stevens CH. Glia: initiators and progressors of pathology in Parkinson's disease. Mov Disord 2011; 26:6-17. [PMID: 21322014 DOI: 10.1002/mds.23455] [Citation(s) in RCA: 311] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
BACKGROUND Glia are traditionally known as support cells for neurons, and their role in neurodegeneration has been largely considered secondary to neuronal dysfunction. We review newer concepts on glial function and assess glial changes in Parkinson's disease (PD) at the time of disease initiation when α-synuclein is accumulating in brain tissue but there is limited neuronal loss, and also as the disease progresses and neuronal loss is evident. RESULTS Of the two main types of astrocytes, only protoplasmic astrocytes are involved in PD, where they become nonreactive and accumulate α-synuclein. Experimental evidence has shown that astrocytic α-synuclein deposition initiates the noncell autonomous killing of neurons through microglial signaling. As the disease progresses, more protoplasmic astrocytes are affected by the disease with an increasing microglial response. Although there is still controversy on the role microglia play in neurodegeneration, there is evidence that microglia are activated early in PD and possibly assist with the clearance of extracellular α-synuclein at this time. Microglia transform to phagocytes and target neurons as the disease progresses but appear to become dysfunctional with increasing amounts of ingested debris. Only nonmyelinating oligodendroglial cells are affected in PD, and only late in the disease process. CONCLUSIONS Glial cells are responsible for the progression of PD and play an important role in initiating the early tissue response. In particular, early dysfunction and α-synuclein accumulation in astrocytes causes recruitment of phagocytic microglia that attack selected neurons in restricted brain regions causing the clinical symptoms of PD.
Collapse
|
3605
|
Maitre M, Roullot-Lacarrière V, Piazza PV, Revest JM. Western blot detection of brain phosphoproteins after performing Laser Microdissection and Pressure Catapulting (LMPC). J Neurosci Methods 2011; 198:204-12. [DOI: 10.1016/j.jneumeth.2011.04.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2010] [Revised: 02/23/2011] [Accepted: 04/01/2011] [Indexed: 01/31/2023]
|
3606
|
Glial dysfunction in the pathogenesis of α-synucleinopathies: emerging concepts. Acta Neuropathol 2011; 121:675-93. [PMID: 21562886 DOI: 10.1007/s00401-011-0833-z] [Citation(s) in RCA: 125] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2011] [Revised: 05/04/2011] [Accepted: 05/04/2011] [Indexed: 12/18/2022]
Abstract
Parkinson's disease (PD), dementia with Lewy bodies (DLB) and multiple system atrophy (MSA) are adult onset neurodegenerative disorders characterised by prominent intracellular α-synuclein aggregates (α-synucleinopathies). The glial contribution to neurodegeneration in α-synucleinopathies was largely underestimated until recently. However, brains of PD and DLB patients exhibit not only neuronal inclusions such as Lewy bodies or Lewy neurites but also glial α-synuclein aggregates. Accumulating experimental evidence in PD models suggests that astrogliosis and microgliosis act as important mediators of neurodegeneration playing a pivotal role in both disease initiation and progression. In MSA, oligodendrocytes are intriguingly affected by aberrant cytoplasmic accumulation of α-synuclein (glial cytoplasmic inclusions, Papp-Lantos bodies). Converging evidence from human postmortem studies and transgenic MSA models suggests that oligodendroglial dysfunction both triggers and exacerbates neuronal degeneration. This review summarises the wide range of responsibilities of astroglia, microglia and oligodendroglia in the healthy brain and the changes in glial function associated with ageing. We then provide a critical analysis of the role of glia in α-synucleinopathies including putative mechanisms promoting a chronically diseased glial microenvironment which can lead to detrimental neuronal changes, including cell loss. Finally, major therapeutic strategies targeting glial pathology in α-synucleinopathies as well as current pitfalls for disease-modification in clinical trials are discussed.
Collapse
|
3607
|
Abstract
Encephalopathy by hyperbilirubinemia in infants has been described for decades, but neither the underlying cellular and molecular mechanisms nor the selective pattern of bilirubin deposition in the brain is well understood. The brain is composed of highly specialized and diverse populations of cells, represented by neurons and glia that comprise astrocytes, oligodendrocytes, and microglia. Although microscopic evaluation of icteric brain sections revealed bilirubin within neurons, neuronal processes, and microglia, cell dependent-sensitivity to bilirubin toxicity and the role of each nerve cell type are poorly understood. Even less considered are glial and neuronal pathologic alterations as integrated phenomena. The available knowledge on reactivity of glial cells to bilirubin and on the impairment to neuronal network dynamics that it causes, here summarized, suggests that a better comprehension of the interplay between neurons and glia is essential to understand bilirubin neurotoxicity and highlight potential molecular targets that may lead to disease-modifying therapeutic approaches.
Collapse
Affiliation(s)
- Dora Brites
- Research Institute for Medicines and Pharmaceutical Sciences (iMedUL), Faculty of Pharmacy, University of Lisbon, Lisbon, Portugal.
| |
Collapse
|
3608
|
CPEB1 modulates lipopolysaccharide-mediated iNOS induction in rat primary astrocytes. Biochem Biophys Res Commun 2011; 409:687-92. [DOI: 10.1016/j.bbrc.2011.05.065] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2011] [Accepted: 05/11/2011] [Indexed: 11/17/2022]
|
3609
|
Bernal GM, Peterson DA. Phenotypic and gene expression modification with normal brain aging in GFAP-positive astrocytes and neural stem cells. Aging Cell 2011; 10:466-82. [PMID: 21385309 DOI: 10.1111/j.1474-9726.2011.00694.x] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Astrocytes secrete growth factors that are both neuroprotective and supportive for the local environment. Identified by glial fibrillary acidic protein (GFAP) expression, astrocytes exhibit heterogeneity in morphology and in the expression of phenotypic markers and growth factors throughout different adult brain regions. In adult neurogenic niches, astrocytes secrete vascular endothelial growth factor (VEGF) and fibroblast growth factor-2 (FGF-2) within the neurogenic niche and are also a source of special GFAP-positive multipotent neural stem cells (NSCs). Normal aging is accompanied by a decline in CNS function and reduced neurogenesis. We asked whether a decreased availability of astrocyte-derived factors may contribute to the age-related decline in neurogenesis. Determining alterations of astrocytic activity in the aging brain is crucial for understanding CNS homeostasis in aging and for assessing appropriate therapeutic targets for an aging population. We found region-specific alterations in the gene expression of GFAP, VEGF, and FGF-2 and their receptors in the aged brain corresponding to changes in astrocytic reactivity, supporting astrocytic heterogeneity and demonstrating a differential aging effect. We found that GFAP-positive NSCs uniquely coexpress both VEGF and its key mitotic receptor Flk-1 in both young and aged hippocampus, indicating a possible autocrine/paracrine signaling mechanism. VEGF expression is lost once NSCs commit to a neuronal fate, but Flk-1-mediated sensitivity to VEGF signaling is maintained. We propose that age-related astrocytic changes result in reduced VEGF and FGF-2 signaling, which in turn limits NSC and progenitor cell maintenance and contributes to decreased neurogenesis.
Collapse
Affiliation(s)
- Giovanna M Bernal
- Department of Neuroscience, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, IL 60647, USA
| | | |
Collapse
|
3610
|
Su Z, Yuan Y, Chen J, Zhu Y, Qiu Y, Zhu F, Huang A, He C. Reactive astrocytes inhibit the survival and differentiation of oligodendrocyte precursor cells by secreted TNF-α. J Neurotrauma 2011; 28:1089-100. [PMID: 21309692 DOI: 10.1089/neu.2010.1597] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Axonal demyelination is a consistent pathological characteristic of spinal cord injury (SCI). Although an increased number of oligodendrocyte progenitor cells (OPCs) is observed in the injured spinal cord, they fail to convert into mature oligodendrocytes. However, little is known about the underlying mechanism. In our study, we identified a link between inhibition of OPC survival and differentiation and reactive astrocytes in glial scar that was mediated by tumor necrosis factor-α (TNF-α). Initially, both glial scar tissue and reactive astrocyte-conditioned medium were shown to inhibit OPC differentiation. Reverse transcriptase polymerase chain reaction (RT-PCR) and immunochemistry revealed that OPCs expressed type 1 TNF-α receptor (TNF-R1). When TNF-α or TNF-R1 was neutralized with antibody, the effect of reactive astrocyte-conditioned medium or recombinant TNF-α protein on OPC differentiation was markedly attenuated. In addition, reactive astrocyte-conditioned medium was also shown to induce OPC apoptosis. All these findings provide the first evidence that reactive astrocytes release TNF-α to inhibit OPC survival and prevent them from differentiating into mature oligodendrocytes, suggesting a mechanism for the failure of remyelination after SCI.
Collapse
Affiliation(s)
- Zhida Su
- Institute of Neuroscience and MOE Key Laboratory of Molecular Neurobiology, Neuroscience Research Center of Changzheng Hospital, Second Military Medical University, Shanghai, China
| | | | | | | | | | | | | | | |
Collapse
|
3611
|
Cengiz P, Kleman N, Uluc K, Kendigelen P, Hagemann T, Akture E, Messing A, Ferrazzano P, Sun D. Inhibition of Na+/H+ exchanger isoform 1 is neuroprotective in neonatal hypoxic ischemic brain injury. Antioxid Redox Signal 2011; 14:1803-13. [PMID: 20712402 PMCID: PMC3078509 DOI: 10.1089/ars.2010.3468] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
We investigated the role of Na(+)/H(+) exchanger isoform 1 (NHE-1) in neonatal hypoxia/ischemia (HI). HI was induced by unilateral ligation of the left common carotid artery in postnatal day 9 (P9) mice, and subsequent exposure of animals to 8% O(2) for 55 min. A pre/posttreatment group received a selective and potent NHE-1 inhibitor HOE 642 (0.5 mg/kg, intraperitoneally) 5 min before HI, then at 24 and 48 h after HI. A posttreatment group received HOE 642 (0.5 mg/kg) at 10 min, 24 h, and 48 h after HI. Saline injections were used as vehicle controls. The vehicle-control brains at 72 h after HI exhibited neuronal degeneration in the ipsilateral hippocampus, striatum, and thalamus, as identified with Fluoro-Jade C positive staining and loss of microtubule-associated protein 2 (MAP2) expression. NHE-1 protein was upregulated in glial fibrillary acidic protein-positive reactive astrocytes. In HOE 642-treated brains, the morphologic hippocampal structures were better preserved and displayed less neurodegeneration and a higher level of MAP2 expression. Motor-learning deficit was detected at 4 weeks of age after HI in the vehicle control group. Inhibition of NHE-1 in P9 mice not only reduced neurodegeneration during the acute stage of HI but also improved the striatum-dependent motor learning and spatial learning at 8 weeks of age after HI. These findings suggest that NHE-1-mediated disruption of ionic homeostasis contributes to striatal and CA1 pyramidal neuronal injury after neonatal HI.
Collapse
Affiliation(s)
- Pelin Cengiz
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
3612
|
Quintas C, Fraga S, Gonçalves J, Queiroz G. P2Y receptors on astrocytes and microglia mediate opposite effects in astroglial proliferation. Purinergic Signal 2011; 7:251-63. [PMID: 21559785 DOI: 10.1007/s11302-011-9235-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2011] [Accepted: 04/28/2011] [Indexed: 01/08/2023] Open
Abstract
Nucleotides released upon brain injury signal to astrocytes and microglia playing an important role in astrogliosis, but the participation of microglia in the purinergic modulation of astrogliosis is still unclear. Highly enriched astroglial cultures and co-cultures of astrocytes and microglia were used to investigate the influence of microglia in the modulation of astroglial proliferation mediated by nucleotides. In highly enriched astroglial cultures, adenosine-5'-triphosphate (ATP), adenosine 5'-O-(3-thio)-triphosphate (ATPγS), adenosine 5'-O-(3-thio)-diphosphate (ADPβS; 0.01-1 mM), and adenosine-5'-diphosphate (ADP; 0.1-1 mM) increased proliferation up to 382%, an effect abolished in co-cultures containing 8% of microglia. The loss of ATP proliferative effect in co-cultures is supported by its fast metabolism and reduced ADP accumulation, an agonist of P2Y(1,12) receptors that mediate astroglial proliferation. No differences in ADPβS and ATPγS metabolism or P2Y(1,12) receptors expression were found in co-cultures that could explain the loss of their proliferative effect. However, conditioned medium from microglia cultures or co-cultures treated with ADPβS, when tested in highly enriched astroglial cultures, also prevented ADPβS proliferative effect. None of the uracil nucleotides tested had any effect in proliferation of highly enriched astroglial cultures, but uridine-5'-triphosphate (UTP; 0.1-1 mM) inhibited proliferation up to 66% in co-cultures, an effect that was dependent on uridine-5'-diphosphate (UDP) accumulation, coincident with a co-localization of P2Y(6) receptors in microglia and due to cell apoptosis. The results indicate that microglia control astroglial proliferation by preventing the proliferative response to adenine nucleotides and favouring an inhibitory effect of UTP/UDP. Several microglial P2Y receptors may be involved by inducing the release of messengers that restrain astrogliosis, a beneficial effect for neuronal repair mechanisms following brain injury.
Collapse
Affiliation(s)
- Clara Quintas
- Laboratory of Pharmacology, Department of Drug Sciences, REQUIMTE, Faculty of Pharmacy, University of Porto, Rua Aníbal Cunha 164, 4050-047, Porto, Portugal
| | | | | | | |
Collapse
|
3613
|
He D, Luo X, Wei W, Xie M, Wang W, Yu Z. DCPIB, A Specific Inhibitor of Volume-Regulated Anion Channels (VRACs), Inhibits Astrocyte Proliferation and Cell Cycle Progression Via G1/S Arrest. J Mol Neurosci 2011; 46:249-57. [DOI: 10.1007/s12031-011-9524-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2011] [Accepted: 04/18/2011] [Indexed: 10/18/2022]
|
3614
|
Holen T. The ultrastructure of lamellar stack astrocytes. Glia 2011; 59:1075-83. [PMID: 21544868 DOI: 10.1002/glia.21180] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2011] [Accepted: 03/29/2011] [Indexed: 11/09/2022]
Abstract
Astrocytes support neurons and map out nonoverlapping domains in grey matter of the brain. The astrocytes of the glia limitans, however, do overlap. Using ultrastructural tools and immunogold histochemistry a subtype of astrocyte able to assemble large lamellar stacks was investigated at the ventral surface of the brain near the hypothalamus. Lamellar stacks were subsequently discovered also in the internal glia limitans of the epithalamus. Circular lamellar stacks containing AQP4 water channels surround neuronal processes, and might serve as osmosensors. The lamellar stacks are well-organized and can form over 100 membrane layers between neuropil and the basal membrane, but a barrier function is not obvious from the noncontinuous character of the stacks along the glia limitans.
Collapse
Affiliation(s)
- Torgeir Holen
- Department of Anatomy, Institute for Basic Medical Science, Sognsvannsveien 9, University of Oslo, 0317 Oslo, Norway.
| |
Collapse
|
3615
|
Neuron-glia signaling: Implications for astrocyte differentiation and synapse formation. Life Sci 2011; 89:524-31. [PMID: 21569780 DOI: 10.1016/j.lfs.2011.04.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2010] [Revised: 03/10/2011] [Accepted: 04/09/2011] [Indexed: 11/22/2022]
Abstract
Glial cells are currently viewed as active partners of neurons in synapse formation. The close proximity of astrocytes to the synaptic cleft implicates that they strongly influence synapse function as well as suggests that these cells might be potential targets for neuronal-released molecules. In this review, we discuss the signaling pathways of astrocyte generation and the role of astrocyte-derived molecules in synapse formation in the central nervous system. Further, we discuss the role of the excitatory neurotransmitter, glutamate and transforming growth factor beta 1 (TGF-β1) pathway in astrocyte generation and differentiation. We provide evidence that astrocytes surrounding synapses are target of neuronal activity and shed light into the role of astroglial cells into neurological disorders associated with glutamate neurotoxicity.
Collapse
|
3616
|
Haroon F, Drögemüller K, Händel U, Brunn A, Reinhold D, Nishanth G, Mueller W, Trautwein C, Ernst M, Deckert M, Schlüter D. Gp130-Dependent Astrocytic Survival Is Critical for the Control of Autoimmune Central Nervous System Inflammation. THE JOURNAL OF IMMUNOLOGY 2011; 186:6521-31. [DOI: 10.4049/jimmunol.1001135] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
3617
|
Philips T, Robberecht W. Neuroinflammation in amyotrophic lateral sclerosis: role of glial activation in motor neuron disease. Lancet Neurol 2011; 10:253-63. [PMID: 21349440 DOI: 10.1016/s1474-4422(11)70015-1] [Citation(s) in RCA: 486] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis (ALS) are characterised by the appearance of reactive microglial and astroglial cells, a process referred to as neuroinflammation. In transgenic mouse models of mutant SOD1-associated familial ALS, reactive microglial cells and astrocytes actively contribute to the death of motor neurons. The biological processes that drive this glial reaction are complex and have both beneficial and deleterious effects on motor neurons. Therapeutic interventions targeting these cells are being explored. An improved understanding of the biological processes that cause neuroinflammation will help to define its medical importance and to identify the therapeutic potential of interfering with this reaction.
Collapse
|
3618
|
Scranton RA, Fletcher L, Sprague S, Jimenez DF, Digicaylioglu M. The rostral migratory stream plays a key role in intranasal delivery of drugs into the CNS. PLoS One 2011; 6:e18711. [PMID: 21533252 PMCID: PMC3076435 DOI: 10.1371/journal.pone.0018711] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2010] [Accepted: 03/16/2011] [Indexed: 01/19/2023] Open
Abstract
Background The blood brain barrier (BBB) is impermeable to most drugs, impeding the establishment of novel neuroprotective therapies and strategies for many neurological diseases. Intranasal administration offers an alternative path for efficient drug delivery into the CNS. So far, the anatomical structures discussed to be involved in the transport of intranasally administered drugs into the CNS include the trigeminal nerve, olfactory nerve and the rostral migratory stream (RMS), but the relative contributions are debated. Methods and Findings In the present study we demonstrate that surgical transection, and the resulting structural disruption of the RMS, in mice effectively obstructs the uptake of intranasally administered radioligands into the CNS. Furthermore, using a fluorescent cell tracer, we demonstrate that intranasal administration in mice allows agents to be distributed throughout the entire brain, including olfactory bulb, hippocampus, cortex and cerebellum. Conclusions This study provides evidence of the vital role the RMS has in the CNS delivery of intranasally administered agents. The identification of the RMS as the major access path for intranasally administered drugs into the CNS may contribute to the development of treatments that are tailored for efficient transport within this structure. Research into the RMS needs to continue to elucidate its limitations, capabilities, mechanisms of transport and potential hazards before we are able to advance this technique into human research.
Collapse
Affiliation(s)
- Robert A. Scranton
- University of Texas Health Science Center San Antonio, San Antonio, Texas, United States of America
| | - Lauren Fletcher
- University of Texas Health Science Center San Antonio, San Antonio, Texas, United States of America
| | - Shane Sprague
- University of Texas Health Science Center San Antonio, San Antonio, Texas, United States of America
| | - David F. Jimenez
- University of Texas Health Science Center San Antonio, San Antonio, Texas, United States of America
| | - Murat Digicaylioglu
- University of Texas Health Science Center San Antonio, San Antonio, Texas, United States of America
- * E-mail:
| |
Collapse
|
3619
|
Geppert M, Hohnholt MC, Thiel K, Nürnberger S, Grunwald I, Rezwan K, Dringen R. Uptake of dimercaptosuccinate-coated magnetic iron oxide nanoparticles by cultured brain astrocytes. NANOTECHNOLOGY 2011; 22:145101. [PMID: 21346306 DOI: 10.1088/0957-4484/22/14/145101] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Magnetic iron oxide nanoparticles (Fe-NP) are currently considered for various diagnostic and therapeutic applications in the brain. However, little is known on the accumulation and biocompatibility of such particles in brain cells. We have synthesized and characterized dimercaptosuccinic acid (DMSA) coated Fe-NP and have investigated their uptake by cultured brain astrocytes. DMSA-coated Fe-NP that were dispersed in physiological medium had an average hydrodynamic diameter of about 60 nm. Incubation of cultured astrocytes with these Fe-NP caused a time- and concentration-dependent accumulation of cellular iron, but did not lead within 6 h to any cell toxicity. After 4 h of incubation with 100-4000 µM iron supplied as Fe-NP, the cellular iron content reached levels between 200 and 2000 nmol mg⁻¹ protein. The cellular iron content after exposure of astrocytes to Fe-NP at 4 °C was drastically lowered compared to cells that had been incubated at 37 °C. Electron microscopy revealed the presence of Fe-NP-containing vesicles in cells that were incubated with Fe-NP at 37 °C, but not in cells exposed to the nanoparticles at 4 °C. These data demonstrate that cultured astrocytes efficiently take up DMSA-coated Fe-NP in a process that appears to be saturable and strongly depends on the incubation temperature.
Collapse
Affiliation(s)
- Mark Geppert
- Center for Biomolecular Interactions Bremen, University of Bremen, PO Box 330440, D-28334 Bremen, Germany
| | | | | | | | | | | | | |
Collapse
|
3620
|
Armentano M, Canalia N, Crociara P, Bonfanti L. Culturing conditions remarkably affect viability and organization of mouse subventricular zone in ex vivo cultured forebrain slices. J Neurosci Methods 2011; 197:65-81. [DOI: 10.1016/j.jneumeth.2011.01.031] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2010] [Revised: 12/29/2010] [Accepted: 01/29/2011] [Indexed: 10/18/2022]
|
3621
|
Pifarre P, Prado J, Baltrons MA, Giralt M, Gabarro P, Feinstein DL, Hidalgo J, Garcia A. Sildenafil (Viagra) ameliorates clinical symptoms and neuropathology in a mouse model of multiple sclerosis. Acta Neuropathol 2011; 121:499-508. [PMID: 21234581 DOI: 10.1007/s00401-010-0795-6] [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] [Received: 10/29/2010] [Revised: 12/21/2010] [Accepted: 12/30/2010] [Indexed: 12/14/2022]
Abstract
Cyclic GMP (cGMP)-mediated pathways regulate inflammatory responses in immune and CNS cells. Recently, cGMP phosphodiesterase inhibitors such as sildenafil, commonly used to treat sexual dysfunction in humans including multiple sclerosis (MS) patients, have been reported to be neuroprotective in animal models of stroke, Alzheimer's disease, and focal brain lesion. In this work, we have examined if sildenafil ameliorates myelin oligodendrocyte glycoprotein peptide (MOG₃₅₋₅₅)-induced experimental autoimmune encephalomyelitis (EAE), a mouse model of MS. We show for the first time that treatment with sildenafil after disease onset markedly reduces the clinical signs of EAE by preventing axonal loss and promoting remyelination. Furthermore, sildenafil decreases CD3+ leukocyte infiltration and microglial/macrophage activation in the spinal cord, while increasing forkhead box transcription factor 3-expressing T regulatory cells (Foxp3 Tregs). However, sildenafil treatment did not significantly affect MOG₃₅₋₅₅-stimulated proliferation or release of Th1/Th2 cytokines in splenocytes but decreased ICAM-1 in spinal cord infiltrated cells. The presence of reactive astrocytes forming scar-like structures around infiltrates was enhanced by sildenafil suggesting a possible mechanism for restriction of leukocyte spread into healthy parenchyma. These results highlight novel actions of sildenafil that may contribute to its beneficial effects in EAE and suggest that treatment with this widely used and well-tolerated drug may be a useful therapeutic intervention to ameliorate MS neuropathology.
Collapse
Affiliation(s)
- Paula Pifarre
- Institute of Biotechnology and Biomedicine, Universitat Autonoma de Barcelona, 08193 Bellaterra, Spain
| | | | | | | | | | | | | | | |
Collapse
|
3622
|
Astroglial inhibition of NF-κB does not ameliorate disease onset and progression in a mouse model for amyotrophic lateral sclerosis (ALS). PLoS One 2011; 6:e17187. [PMID: 21445241 PMCID: PMC3060799 DOI: 10.1371/journal.pone.0017187] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2010] [Accepted: 01/25/2011] [Indexed: 12/14/2022] Open
Abstract
Motor neuron death in amyotrophic lateral sclerosis (ALS) is considered a “non-cell autonomous” process, with astrocytes playing a critical role in disease progression. Glial cells are activated early in transgenic mice expressing mutant SOD1, suggesting that neuroinflammation has a relevant role in the cascade of events that trigger the death of motor neurons. An inflammatory cascade including COX2 expression, secretion of cytokines and release of NO from astrocytes may descend from activation of a NF-κB-mediated pathway observed in astrocytes from ALS patients and in experimental models. We have attempted rescue of transgenic mutant SOD1 mice through the inhibition of the NF-κB pathway selectively in astrocytes. Here we show that despite efficient inhibition of this major pathway, double transgenic mice expressing the mutant SOD1G93A ubiquitously and the dominant negative form of IκBα (IκBαAA) in astrocytes under control of the GFAP promoter show no benefit in terms of onset and progression of disease. Our data indicate that motor neuron death in ALS cannot be prevented by inhibition of a single inflammatory pathway because alternative pathways are activated in the presence of a persistent toxic stimulus.
Collapse
|
3623
|
Quintas C, Fraga S, Gonçalves J, Queiroz G. Opposite modulation of astroglial proliferation by adenosine 5'-O-(2-thio)-diphosphate and 2-methylthioadenosine-5'-diphosphate: mechanisms involved. Neuroscience 2011; 182:32-42. [PMID: 21419195 DOI: 10.1016/j.neuroscience.2011.03.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2010] [Revised: 03/01/2011] [Accepted: 03/02/2011] [Indexed: 01/26/2023]
Abstract
The contribution of P2Y(12,13) receptors to astroglial proliferation was investigated by testing the effects of two agonists with high affinity for these receptors, adenosine 5'-O-(2-thio)-diphosphate (ADPβS) and 2-methylthioadenosine-5'-diphosphate (2-MeSADP), in the incorporation of [(3)H]-thymidine. The effect of ATP, an endogenous inducer of astroglial proliferation, was also investigated. ADPβS and ATP (0.01-1 mM) increased astroglial proliferation up to 282%, an effect inhibited by the P2Y(1) receptor antagonist MRS 2179 (30 μM). The P2Y(12) receptor antagonists MRS 2395 (10 μM) and AR-C 66096 (10 μM) also reduced ADPβS proliferative effect, whereas the effect of ATP was attenuated by the A(2A) and A(2B) receptor antagonists SCH 58261 (30 nM) and MRS 1706 (10 nM), respectively. Studies of the signalling pathway activated showed that ADPβS effect was attenuated by pertussis toxin and by inhibition of phopholipase C (PLC), protein kinase C (PKC) and extracellular signal-regulated kinase1/2 (ERK1/2). The effect of ATP was also attenuated by inhibition of protein kinase A (PKA). The agonist 2-MeSADP (0.001-10 μM) had no effect in astroglial proliferation, but at higher concentrations (0.1-1 mM) it inhibited up to 63%, by mechanisms independent of P2Y(1,12,13) receptors activation. It was metabolised into 2-methylthioadenosine (2-MeSADO), the metabolite responsible for inhibition of astroglial proliferation. The effect of 2-MeSADO (0.1 mM) was attenuated by the A(3) receptors antagonist MRS 1523 (10 μM) and by the inhibitor of nucleoside transporters uridine (0.3 mM). 2-MeSADO did not induce apoptosis but increased lactate dehydrogenase release, an indicator of necrotic cell death. Astroglial proliferation induced by ADPβS was mediated by P2Y(1) and P2Y(12) receptors, leading to activation of PLC-PKC-ERK1/2 signalling pathway. The ATP proliferative effect was also mediated by PKA, supporting the contribution of the A(2) receptors. 2-MeSADP inhibition of astroglial proliferation depended on its conversion into 2-MeSADO, which activated A(3) receptors, blocked [(3)H]-thymidine uptake by astrocytes and led to cell death.
Collapse
Affiliation(s)
- C Quintas
- Department of Drug Sciences, Faculty of Pharmacy, Centro de Química Fina e Biotecnologia da Universidade de Lisboa e Centro de Química da Universidade do Porto (REQUIMTE), University of Porto, Porto, Portugal
| | | | | | | |
Collapse
|
3624
|
Miraucourt LS, Peirs C, Dallel R, Voisin DL. Glycine inhibitory dysfunction turns touch into pain through astrocyte-derived D-serine. Pain 2011; 152:1340-1348. [PMID: 21392888 DOI: 10.1016/j.pain.2011.02.021] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2010] [Revised: 01/13/2011] [Accepted: 02/07/2011] [Indexed: 12/26/2022]
Abstract
Glycine inhibitory dysfunction provides a useful experimental model for studying the mechanism of dynamic mechanical allodynia, a widespread and intractable symptom of neuropathic pain. In this model, allodynia expression relies on N-methyl-d-aspartate receptors (NMDARs), and it has been shown that astrocytes can regulate their activation through the release of the NMDAR coagonist d-serine. Recent studies also suggest that astrocytes potentially contribute to neuropathic pain. However, the involvement of astrocytes in dynamic mechanical allodynia remains unknown. Here, we show that after blockade of glycine inhibition, orofacial tactile stimuli activated medullary dorsal horn (MDH) astrocytes, but not microglia. Accordingly, the glia inhibitor fluorocitrate, but not the microglia inhibitor minocycline, prevented allodynia. Fluorocitrate also impeded activation of astrocytes and blocked activation of the superficial MDH neural circuit underlying allodynia, as revealed by study of Fos expression. MDH astrocytes are thus required for allodynia. They may also produce d-serine because astrocytic processes were selectively immunolabeled for serine racemase, the d-serine synthesizing enzyme. Accordingly, selective degradation of d-serine with d-amino acid oxidase applied in vivo prevented allodynia and activation of the underlying neural circuit. Conversely, allodynia blockade by fluorocitrate was reversed by exogenous d-serine. These results suggest the following scenario: removal of glycine inhibition makes tactile stimuli able to activate astrocytes; activated astrocytes may provide d-serine to enable NMDAR activation and thus allodynia. Such a contribution of astrocytes to pathological pain fuels the emerging concept that astrocytes are critical players in pain signaling. Glycine disinhibition makes tactile stimuli able to activate astrocytes, which may provide d-serine to enable NMDA receptor activation and thus allodynia.
Collapse
Affiliation(s)
- Loïs S Miraucourt
- Clermont Université, Université d'Auvergne, Neurobiologie de la douleur trigéminale, BP 10448, F-63000 Clermont-Ferrand, France Inserm, U929, F-63000 Clermont-Ferrand, CHU Clermont-Ferrand, Service d'Odontologie, F-63003 Clermont-Ferrand, France Inserm, U862, Neurocentre Magendie, F-33077 Bordeaux, France Université de Bordeaux, F-33077 Bordeaux, France
| | | | | | | |
Collapse
|
3625
|
Tsuda M, Kohro Y, Yano T, Tsujikawa T, Kitano J, Tozaki-Saitoh H, Koyanagi S, Ohdo S, Ji RR, Salter MW, Inoue K. JAK-STAT3 pathway regulates spinal astrocyte proliferation and neuropathic pain maintenance in rats. ACTA ACUST UNITED AC 2011; 134:1127-39. [PMID: 21371995 DOI: 10.1093/brain/awr025] [Citation(s) in RCA: 247] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Neuropathic pain, a debilitating pain condition, is a common consequence of damage to the nervous system. Optimal treatment of neuropathic pain is a major clinical challenge because the underlying mechanisms remain unclear and currently available treatments are frequently ineffective. Emerging lines of evidence indicate that peripheral nerve injury converts resting spinal cord glia into reactive cells that are required for the development and maintenance of neuropathic pain. However, the mechanisms underlying reactive astrogliosis after nerve injury are largely unknown. In the present study, we investigated cell proliferation, a critical process in reactive astrogliosis, and determined the temporally restricted proliferation of dorsal horn astrocytes in rats with spinal nerve injury, a well-known model of neuropathic pain. We found that nerve injury-induced astrocyte proliferation requires the Janus kinase-signal transducers and activators of transcription 3 signalling pathway. Nerve injury induced a marked signal transducers and activators of transcription 3 nuclear translocation, a primary index of signal transducers and activators of transcription 3 activation, in dorsal horn astrocytes. Intrathecally administering inhibitors of Janus kinase-signal transducers and activators of transcription 3 signalling to rats with nerve injury reduced the number of proliferating dorsal horn astrocytes and produced a recovery from established tactile allodynia, a cardinal symptom of neuropathic pain that is characterized by pain hypersensitivity evoked by innocuous stimuli. Moreover, recovery from tactile allodynia was also produced by direct suppression of dividing astrocytes by intrathecal administration of the cell cycle inhibitor flavopiridol to nerve-injured rats. Together, these results imply that the Janus kinase-signal transducers and activators of transcription 3 signalling pathway are critical transducers of astrocyte proliferation and maintenance of tactile allodynia and may be a therapeutic target for neuropathic pain.
Collapse
Affiliation(s)
- Makoto Tsuda
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, Fukuoka 812-8582, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
3626
|
Takado Y, Igarashi H, Terajima K, Shimohata T, Ozawa T, Okamoto K, Nishizawa M, Nakada T. Brainstem metabolites in multiple system atrophy of cerebellar type: 3.0-T magnetic resonance spectroscopy study. Mov Disord 2011; 26:1297-302. [PMID: 21370263 DOI: 10.1002/mds.23550] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2010] [Revised: 10/13/2010] [Accepted: 11/01/2010] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND The aim of this study was to find biomarkers of disease severity in multiple system atrophy of cerebellar type by imaging disease specific regions using proton magnetic resonance spectroscopy on a 3.0 T system. METHODS We performed proton magnetic resonance spectroscopy separately in the pons and medulla on 12 multiple system atrophy of cerebellar type patients and 12 age and gender matched control subjects. The metabolite concentrations were estimated from single-voxel proton magnetic resonance spectra measured by point resolved spectroscopy, which were then correlated with clinical severity using Part I, II, and IV of the unified multiple system atrophy rating scale. RESULTS Proton magnetic resonance spectroscopy showed that myo-inositol concentrations in both the pons and medulla were significantly higher in multiple system atrophy of cerebellar type patients compared to those of the control subjects (P < 0.05). By contrast, total N-acetylaspartate (the sum of N-acetylaspartate and N-acetylaspartylglutamate) and total choline compounds concentrations in both the pons and medulla were significantly lower in multiple system atrophy of cerebellar type patients compared to control subjects (P < 0.05). Creatine concentration in the pons was significantly higher in multiple system atrophy of cerebellar type patients compared to the control subjects (P < 0.05). Furthermore, a significant correlation was found between the myo-inositol/creatine ratio in the pons and clinical severity, defined by the sum score of unified multiple system atrophy rating scale (I+II+IV) (r = 0.76, P < 0.01). CONCLUSION Proton magnetic resonance spectroscopy, in conjunction with a 3.0 T system, can be feasible to detect part of pathological changes in the brainstem, such as gliosis and neuronal cell loss, and the metabolites can be used as biomarkers of clinical severity in multiple system atrophy of cerebellar type patients.
Collapse
Affiliation(s)
- Yuhei Takado
- Center for Integrated Human Brain Science, Brain Research Institute, University of Niigata, Niigata, Japan
| | | | | | | | | | | | | | | |
Collapse
|
3627
|
Li W, Poteet E, Xie L, Liu R, Wen Y, Yang SH. Regulation of matrix metalloproteinase 2 by oligomeric amyloid β protein. Brain Res 2011; 1387:141-8. [PMID: 21376707 DOI: 10.1016/j.brainres.2011.02.078] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2010] [Revised: 02/22/2011] [Accepted: 02/24/2011] [Indexed: 01/08/2023]
Abstract
Matrix metalloproteinases (MMPs) are a group of proteinases that degrade components of the extracellular matrix (ECM). There is increasing evidence for a link between the activation of MMPs and Alzheimer's disease (AD) pathogenesis, in which both beneficial and detrimental actions of MMPs have been suggested. It has been demonstrated that MMPs could degrade amyloid β (Aβ) and play important roles in the extracellular Aβ catabolism and clearance. On the other hand, MMPs could contribute to AD pathogenesis by compromising the blood brain barrier and promoting neurodegeneration. In the present study, we observed that oligomeric Aβ regulates MMP2 expression in a paradoxical manner. In rat primary astrocyte cultures, oligomeric Aβ down-regulated MMP2 transcription and reduced its extracellular activity. However, in a widely used mouse model for AD, immunohistochemistry demonstrated an increase of MMP2 expression in astrocytes surrounding senile plaques in APP/PS1 transgenic mice brains. Using real-time PCR, we found that the MMP2 mRNA level was elevated in APP/PS1 transgenic mice brains. In addition, elevated mRNA levels of MMP stimulating cytokines such as IL-1β and TGFβ were found in the brains of APP/PS1 mice. Our study suggests a complex regulation of MMP2 expression by oligomeric Aβ in astrocytes. While oligomeric Aβ directly down-regulates MMP2 expression and activation in astrocytes, it induces production of proinflammatory cytokines which could serve as strong stimulators for MMP2. Therefore, the ultimate outcome of the oligomeric Aβ on MMP2 activation in astrocytes might be the combination of its direct inhibitory action on astrocyte MMP2 expression and the secondary action of inducing inflammatory cytokines.
Collapse
Affiliation(s)
- Wenjun Li
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | | | | | | | | | | |
Collapse
|
3628
|
Robel S, Berninger B, Götz M. The stem cell potential of glia: lessons from reactive gliosis. Nat Rev Neurosci 2011; 12:88-104. [PMID: 21248788 DOI: 10.1038/nrn2978] [Citation(s) in RCA: 394] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Astrocyte-like cells, which act as stem cells in the adult brain, reside in a few restricted stem cell niches. However, following brain injury, glia outside these niches acquire or reactivate stem cell potential as part of reactive gliosis. Recent studies have begun to uncover the molecular pathways involved in this process. A comparison of molecular pathways activated after injury with those involved in the normal neural stem cell niches highlights strategies that could overcome the inhibition of neurogenesis outside the stem cell niche and instruct parenchymal glia towards a neurogenic fate. This new view on reactive glia therefore suggests a widespread endogenous source of cells with stem cell potential, which might potentially be harnessed for local repair strategies.
Collapse
Affiliation(s)
- Stefanie Robel
- Physiological Genomics, Ludwig-Maximilians University of Munich, Germany
| | | | | |
Collapse
|
3629
|
Hashioka S, Klegeris A, McGeer PL. Proton pump inhibitors reduce interferon-γ-induced neurotoxicity and STAT3 phosphorylation of human astrocytes. Glia 2011; 59:833-40. [PMID: 21360757 DOI: 10.1002/glia.21157] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2010] [Accepted: 01/19/2011] [Indexed: 01/12/2023]
Abstract
Proton pump inhibitors (PPIs) are known to possess anti-inflammatory properties. Inflammatory processes, including astrocytic activation, are implicated in the pathogenesis of different neurodegenerative diseases. Our recent study has indicated that interferon (IFN)-γ-induced astrocytic neurotoxicity is mediated, at least in part, by phosphorylation of signal transducer and activator of transcription (STAT) 3. We therefore studied the effects of PPIs on IFN-γ-induced neurotoxicity and STAT3 activation of human astrocytes. Both lansoprazole (LPZ) and omeprazole (OPZ) significantly attenuated IFN-γ-induced neurotoxicity of human astrocytes and astrocytoma cells. These drugs inhibited IFN-γ-induced phosphorylation of STAT 3, but not STAT1. We found that LPZ significantly reduced secretion of IFN-γ-inducible T cell α chemoattractant from IFN-γ-activated astrocytes. Neither LPZ nor OPZ suppressed expression of intercellular adhesion molecule-1 by IFN-γ-activated astrocytes. These results suggest that PPIs attenuate IFN-γ-induced neurotoxicity of human astrocytes through inhibition of the STAT3 signaling pathway. PPIs that possess antineurotoxic properties may be a useful treatment option for Alzheimer's disease and other neuroinflammatory disorders associated with activated astrocytes.
Collapse
Affiliation(s)
- Sadayuki Hashioka
- Department of Psychiatry, The University of British Columbia, Kinsmen Laboratory of Neurological Research, Vancouver, B.C., Canada
| | | | | |
Collapse
|
3630
|
L’Episcopo F, Tirolo C, Testa N, Caniglia S, Morale M, Cossetti C, D’Adamo P, Zardini E, Andreoni L, Ihekwaba A, Serra P, Franciotta D, Martino G, Pluchino S, Marchetti B. Reactive astrocytes and Wnt/β-catenin signaling link nigrostriatal injury to repair in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine model of Parkinson's disease. Neurobiol Dis 2011; 41:508-27. [PMID: 21056667 PMCID: PMC3558878 DOI: 10.1016/j.nbd.2010.10.023] [Citation(s) in RCA: 160] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2010] [Revised: 10/15/2010] [Accepted: 10/27/2010] [Indexed: 02/07/2023] Open
Abstract
Emerging evidence points to reactive glia as a pivotal factor in Parkinson's disease (PD) and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-lesioned mouse model of basal ganglia injury, but whether astrocytes and microglia activation may exacerbate dopaminergic (DAergic) neuron demise and/or contribute to DAergic repair is presently the subject of much debate. Here, we have correlated the loss and recovery of the nigrostriatal DAergic functionality upon acute MPTP exposure with extensive gene expression analysis at the level of the ventral midbrain (VM) and striata (Str) and found a major upregulation of pro-inflammatory chemokines and wingless-type MMTV integration site1 (Wnt1), a key transcript involved in midbrain DAergic neurodevelopment. Wnt signaling components (including Frizzled-1 [Fzd-1] and β-catenin) were dynamically regulated during MPTP-induced DAergic degeneration and reactive glial activation. Activated astrocytes of the ventral midbrain were identified as candidate source of Wnt1 by in situ hybridization and real-time PCR in vitro. Blocking Wnt/Fzd signaling with Dickkopf-1 (Dkk1) counteracted astrocyte-induced neuroprotection against MPP(+) toxicity in primary mesencephalic astrocyte-neuron cultures, in vitro. Moreover, astroglial-derived factors, including Wnt1, promoted neurogenesis and DAergic neurogenesis from adult midbrain stem/neuroprogenitor cells, in vitro. Conversely, lack of Wnt1 transcription in response to MPTP in middle-aged mice and failure of DAergic neurons to recover were reversed by pharmacological activation of Wnt/β-catenin signaling, in vivo, thus suggesting MPTP-reactive astrocytes in situ and Wnt1 as candidate components of neuroprotective/neurorescue pathways in MPTP-induced nigrostriatal DAergic plasticity.
Collapse
Affiliation(s)
- F. L’Episcopo
- OASI Institute for Research and Care on Mental Retardation and Brain Aging, Neuropharmacology Section, Via Conte Ruggero 73, 94018 Troina (EN) Italy
| | - C. Tirolo
- OASI Institute for Research and Care on Mental Retardation and Brain Aging, Neuropharmacology Section, Via Conte Ruggero 73, 94018 Troina (EN) Italy
| | - N. Testa
- OASI Institute for Research and Care on Mental Retardation and Brain Aging, Neuropharmacology Section, Via Conte Ruggero 73, 94018 Troina (EN) Italy
| | - S. Caniglia
- OASI Institute for Research and Care on Mental Retardation and Brain Aging, Neuropharmacology Section, Via Conte Ruggero 73, 94018 Troina (EN) Italy
| | - M.C. Morale
- OASI Institute for Research and Care on Mental Retardation and Brain Aging, Neuropharmacology Section, Via Conte Ruggero 73, 94018 Troina (EN) Italy
| | - C. Cossetti
- Cambridge Centre for Brain Repair Department of Clinical Neurosciences ED Adrian Building Forvie Site Robinson Way Cambridge CB2 0PY, UK
| | - P. D’Adamo
- Molecular Genetics of Mental Retardation Unit, San Raffaele Institute, Via Olgettina, 58 I-20132 Milan, Italy
| | - E. Zardini
- Neuroimmunology Unit, National Neurological Instititute C. Mondino, Via Mondino 2, 27100 Pavia, Italy
| | - L. Andreoni
- Neuroimmunology Unit, National Neurological Instititute C. Mondino, Via Mondino 2, 27100 Pavia, Italy
| | - A.E.C. Ihekwaba
- Cambridge Centre for Brain Repair Department of Clinical Neurosciences ED Adrian Building Forvie Site Robinson Way Cambridge CB2 0PY, UK
| | - P.A. Serra
- Department of Pharmacology, University of Sassari, Medical School, Viale S. Pietro 43, 07100 Sassari, Italy
| | - D. Franciotta
- Department of Pharmacology, University of Sassari, Medical School, Viale S. Pietro 43, 07100 Sassari, Italy
| | - G. Martino
- San Raffaele Institute, Neuroimmunology Unit, DIBIT2 and Institute of Experimental Neurology (INSPE), Via Olgettina, 58 I-20132 Milan, Italy
| | - S. Pluchino
- Cambridge Centre for Brain Repair Department of Clinical Neurosciences ED Adrian Building Forvie Site Robinson Way Cambridge CB2 0PY, UK
| | - B. Marchetti
- OASI Institute for Research and Care on Mental Retardation and Brain Aging, Neuropharmacology Section, Via Conte Ruggero 73, 94018 Troina (EN) Italy
- Department of Clinical and Molecular Biomedicine, Pharmacology Section, University of Catania, Viale A. Doria, 95125 Catania, Italy
- Faculty of Pharmacy, University of Catania, Viale A. Doria, 95125 Catania, Italy
| |
Collapse
|
3631
|
Lee JH, Yu HS, Lee GS, Ji A, Hyun JK, Kim HW. Collagen gel three-dimensional matrices combined with adhesive proteins stimulate neuronal differentiation of mesenchymal stem cells. J R Soc Interface 2011; 8:998-1010. [PMID: 21247946 DOI: 10.1098/rsif.2010.0613] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Three-dimensional gel matrices provide specialized microenvironments that mimic native tissues and enable stem cells to grow and differentiate into specific cell types. Here, we show that collagen three-dimensional gel matrices prepared in combination with adhesive proteins, such as fibronectin (FN) and laminin (LN), provide significant cues to the differentiation into neuronal lineage of mesenchymal stem cells (MSCs) derived from rat bone marrow. When cultured within either a three-dimensional collagen gel alone or one containing either FN or LN, and free of nerve growth factor (NGF), the MSCs showed the development of numerous neurite outgrowths. These were, however, not readily observed in two-dimensional culture without the use of NGF. Immunofluorescence staining, western blot and fluorescence-activated cell sorting analyses demonstrated that a large population of cells was positive for NeuN and glial fibrillary acidic protein, which are specific to neuronal cells, when cultured in the three-dimensional collagen gel. The dependence of the neuronal differentiation of MSCs on the adhesive proteins containing three-dimensional gel matrices is considered to be closely related to focal adhesion kinase (FAK) activation through integrin receptor binding, as revealed by an experiment showing no neuronal outgrowth in the FAK-knockdown cells and stimulation of integrin β1 gene. The results provided herein suggest the potential role of three-dimensional collagen-based gel matrices combined with adhesive proteins in the neuronal differentiation of MSCs, even without the use of chemical differentiation factors. Furthermore, these findings suggest that three-dimensional gel matrices might be useful as nerve-regenerative scaffolds.
Collapse
Affiliation(s)
- Jae Ho Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, , South Korea
| | | | | | | | | | | |
Collapse
|
3632
|
Tufekci KU, Genc S, Genc K. The endotoxin-induced neuroinflammation model of Parkinson's disease. PARKINSON'S DISEASE 2011; 2011:487450. [PMID: 21331154 PMCID: PMC3034925 DOI: 10.4061/2011/487450] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2010] [Revised: 11/18/2010] [Accepted: 12/16/2010] [Indexed: 01/22/2023]
Abstract
Parkinson's disease (PD) is a common neurodegenerative disorder characterized by the progressive loss of dopaminergic (DA) neurons in the substantia nigra. Although the exact cause of the dopaminergic neurodegeneration remains elusive, recent postmortem and experimental studies have revealed an essential role for neuroinflammation that is initiated and driven by activated microglial and infiltrated peripheral immune cells and their neurotoxic products (such as proinflammatory cytokines, reactive oxygen species, and nitric oxide) in the pathogenesis of PD. A bacterial endotoxin-based experimental model of PD has been established, representing a purely inflammation-driven animal model for the induction of nigrostriatal dopaminergic neurodegeneration. This model, by itself or together with genetic and toxin-based animal models, provides an important tool to delineate the precise mechanisms of neuroinflammation-mediated dopaminergic neuron loss. Here, we review the characteristics of this model and the contribution of neuroinflammatory processes, induced by the in vivo administration of bacterial endotoxin, to neurodegeneration. Furthermore, we summarize the recent experimental therapeutic strategies targeting endotoxin-induced neuroinflammation to elicit neuroprotection in the nigrostriatal dopaminergic system. The potential of the endotoxin-based PD model in the development of an early-stage specific diagnostic biomarker is also emphasized.
Collapse
Affiliation(s)
- Kemal Ugur Tufekci
- Department of Neuroscience, Health Science Institute, Dokuz Eylul University, Inciralti, 35340 Izmir, Turkey
| | - Sermin Genc
- Department of Neuroscience, Health Science Institute, Dokuz Eylul University, Inciralti, 35340 Izmir, Turkey
| | - Kursad Genc
- Department of Neuroscience, Health Science Institute, Dokuz Eylul University, Inciralti, 35340 Izmir, Turkey
| |
Collapse
|
3633
|
Kang W, Hébert JM. Signaling pathways in reactive astrocytes, a genetic perspective. Mol Neurobiol 2011; 43:147-54. [PMID: 21234816 DOI: 10.1007/s12035-011-8163-7] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2010] [Accepted: 01/03/2011] [Indexed: 12/11/2022]
Abstract
Reactive astrocytes are associated with a vast array of central nervous system (CNS) pathologies. The activation of astrocytes is characterized by changes in their molecular and morphological features, and depending on the type of damage can also be accompanied by inflammatory responses, neuronal damage, and in severe cases, scar formation. Although reactive astrogliosis is the normal physiological response essential for containing damage, it can also have detrimental effects on neuronal survival and axon regeneration, particularly in neurodegenerative diseases. It is believed that progressive changes in astrocytes as they become reactive are finely regulated by complex intercellular and intracellular signaling mechanisms. However, these have yet to be sorted out. Much has been learned from gain-of-function approaches in vivo and culture paradigms, but in most cases, loss-of-function genetic studies, which are a critical complementary approach, have been lacking. Understanding which signaling pathways are required to control different aspects of astrogliosis will be necessary for designing therapeutic strategies to improve their beneficial effects and limit their detrimental ones in CNS pathologies. In this article, we review recent advances in the mechanisms underlying the regulation of aspects of astrogliosis, with the main focus on the signaling pathways that have been studied using loss-of-function genetic mouse models.
Collapse
Affiliation(s)
- Wenfei Kang
- Department of Neuroscience and Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | |
Collapse
|
3634
|
Förtsch C, Hupp S, Ma J, Mitchell TJ, Maier E, Benz R, Iliev AI. Changes in astrocyte shape induced by sublytic concentrations of the cholesterol-dependent cytolysin pneumolysin still require pore-forming capacity. Toxins (Basel) 2011; 3:43-62. [PMID: 22069689 PMCID: PMC3210454 DOI: 10.3390/toxins3010043] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2010] [Revised: 12/30/2010] [Accepted: 01/04/2011] [Indexed: 11/21/2022] Open
Abstract
Streptococcus pneumoniae is a common pathogen that causes various infections, such as sepsis and meningitis. A major pathogenic factor of S. pneumoniae is the cholesterol-dependent cytolysin, pneumolysin. It produces cell lysis at high concentrations and apoptosis at lower concentrations. We have shown that sublytic amounts of pneumolysin induce small GTPase-dependent actin cytoskeleton reorganization and microtubule stabilization in human neuroblastoma cells that are manifested by cell retraction and changes in cell shape. In this study, we utilized a live imaging approach to analyze the role of pneumolysin’s pore-forming capacity in the actin-dependent cell shape changes in primary astrocytes. After the initial challenge with the wild-type toxin, a permeabilized cell population was rapidly established within 20-40 minutes. After the initial rapid permeabilization, the size of the permeabilized population remained unchanged and reached a plateau. Thus, we analyzed the non-permeabilized (non-lytic) population, which demonstrated retraction and shape changes that were inhibited by actin depolymerization. Despite the non-lytic nature of pneumolysin treatment, the toxin’s lytic capacity remained critical for the initiation of cell shape changes. The non-lytic pneumolysin mutants W433F-pneumolysin and delta6-pneumolysin, which bind the cell membrane with affinities similar to that of the wild-type toxin, were not able to induce shape changes. The initiation of cell shape changes and cell retraction by the wild-type toxin were independent of calcium and sodium influx and membrane depolarization, which are known to occur following cellular challenge and suggested to result from the ion channel-like properties of the pneumolysin pores. Excluding the major pore-related phenomena as the initiation mechanism of cell shape changes, the existence of a more complex relationship between the pore-forming capacity of pneumolysin and the actin cytoskeleton reorganization is suggested.
Collapse
Affiliation(s)
- Christina Förtsch
- DFG Membrane, Cytoskeleton Interaction Group, Institute of Pharmacology and Toxicology & Rudolf Virchow Center for Experimental Medicine, University of Würzburg, Versbacherstr. 9, 97078 Würzburg, Germany; (C.F.); (S.H.)
| | - Sabrina Hupp
- DFG Membrane, Cytoskeleton Interaction Group, Institute of Pharmacology and Toxicology & Rudolf Virchow Center for Experimental Medicine, University of Würzburg, Versbacherstr. 9, 97078 Würzburg, Germany; (C.F.); (S.H.)
| | - Jiangtao Ma
- Division of Infection and Immunity, Level 2, Glasgow Biomedical Research Centre, University of Glasgow, 120 University Place, Glasgow, G12 8TA, UK; (J.M.); (T.J.M.)
| | - Timothy J. Mitchell
- Division of Infection and Immunity, Level 2, Glasgow Biomedical Research Centre, University of Glasgow, 120 University Place, Glasgow, G12 8TA, UK; (J.M.); (T.J.M.)
| | - Elke Maier
- Rudolf Virchow Center for Experimental Medicine, University of Würzburg, Versbacherstr. 9, 97078 Würzburg, Germany; (E.M.); (R.B.)
| | - Roland Benz
- Rudolf Virchow Center for Experimental Medicine, University of Würzburg, Versbacherstr. 9, 97078 Würzburg, Germany; (E.M.); (R.B.)
| | - Asparouh I. Iliev
- DFG Membrane, Cytoskeleton Interaction Group, Institute of Pharmacology and Toxicology & Rudolf Virchow Center for Experimental Medicine, University of Würzburg, Versbacherstr. 9, 97078 Würzburg, Germany; (C.F.); (S.H.)
- Author to whom correspondence should be addressed; ; Tel.: +49-931-20148997; Fax: +49-931-20148539
| |
Collapse
|
3635
|
Skilled reaching training promotes astroglial changes and facilitated sensorimotor recovery after collagenase-induced intracerebral hemorrhage. Exp Neurol 2011; 227:53-61. [DOI: 10.1016/j.expneurol.2010.09.009] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2010] [Revised: 09/04/2010] [Accepted: 09/08/2010] [Indexed: 11/20/2022]
|
3636
|
Zeng HC, Zhang L, Li YY, Wang YJ, Xia W, Lin Y, Wei J, Xu SQ. Inflammation-like glial response in rat brain induced by prenatal PFOS exposure. Neurotoxicology 2011; 32:130-9. [DOI: 10.1016/j.neuro.2010.10.001] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2010] [Revised: 09/17/2010] [Accepted: 10/05/2010] [Indexed: 11/25/2022]
|
3637
|
Béjot Y, Prigent-Tessier A, Cachia C, Giroud M, Mossiat C, Bertrand N, Garnier P, Marie C. Time-dependent contribution of non neuronal cells to BDNF production after ischemic stroke in rats. Neurochem Int 2011; 58:102-11. [DOI: 10.1016/j.neuint.2010.10.019] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2010] [Revised: 10/26/2010] [Accepted: 10/31/2010] [Indexed: 12/19/2022]
|
3638
|
Blanc F, Martinian L, Liagkouras I, Catarino C, Sisodiya SM, Thom M. Investigation of widespread neocortical pathology associated with hippocampal sclerosis in epilepsy: A postmortem study. Epilepsia 2010; 52:10-21. [DOI: 10.1111/j.1528-1167.2010.02773.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
3639
|
|
3640
|
Panneton WM, Kumar VB, Gan Q, Burke WJ, Galvin JE. The neurotoxicity of DOPAL: behavioral and stereological evidence for its role in Parkinson disease pathogenesis. PLoS One 2010; 5:e15251. [PMID: 21179455 PMCID: PMC3001493 DOI: 10.1371/journal.pone.0015251] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2010] [Accepted: 11/05/2010] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The etiology of Parkinson disease (PD) has yet to be fully elucidated. We examined the consequences of injections of 3,4-dihydroxyphenylacetaldehyde (DOPAL), a toxic metabolite of dopamine, into the substantia nigra of rats on motor behavior and neuronal survival. METHODS/PRINCIPAL FINDINGS A total of 800 nl/rat of DOPAL (1 µg/200 nl) was injected stereotaxically into the substantia nigra over three sites while control animals received similar injections of phosphate buffered saline. Rotational behavior of these rats was analyzed, optical density of striatal tyrosine hydroxylase was calculated, and unbiased stereological counts of the substantia nigra were made. The rats showed significant rotational asymmetry ipsilateral to the lesion, supporting disruption of dopaminergic nigrostriatal projections. Such disruption was verified since the density of striatal tyrosine hydroxylase decreased significantly (p<0.001) on the side ipsilateral to the DOPAL injections when compared to the non-injected side. Stereological counts of neurons stained for Nissl in pars compacta of the substantia nigra significantly decreased (p<0.001) from control values, while counts of those in pars reticulata were unchanged after DOPAL injections. Counts of neurons immunostained for tyrosine hydroxylase also showed a significant (p=0.032) loss of dopaminergic neurons. In spite of significant loss of dopaminergic neurons, DOPAL injections did not induce significant glial reaction in the substantia nigra. CONCLUSIONS The present study provides the first in vivo quantification of substantia nigra pars compacta neuronal loss after injection of the endogenous toxin DOPAL. The results demonstrate that injections of DOPAL selectively kills SN DA neurons, suggests loss of striatal DA terminals, spares non-dopaminergic neurons of the pars reticulata, and triggers a behavioral phenotype (rotational asymmetry) consistent with other PD animal models. This study supports the "catecholaldehyde hypothesis" as an important link for the etiology of sporadic PD.
Collapse
Affiliation(s)
- W Michael Panneton
- Department of Pharmacological and Physiological Science, Saint Louis University School of Medicine, St. Louis, Missouri, United States of America.
| | | | | | | | | |
Collapse
|
3641
|
Abstract
The intent of this article is to assist pathologists inexperienced in examining central nervous system (CNS) sections to recognize normal and abnormal cell types as well as some common artifacts. Dark neurons are the most common histologic artifact but, with experience, can readily be distinguished from degenerating (eosinophilic) neurons. Neuron degeneration stains can be useful in lowering the threshold for detecting neuron degeneration as well as for revealing degeneration within populations of neurons that are too small to show the associated eosinophilic cytoplasmic alteration within H&E-stained sections. Neuron degeneration may also be identified by the presence of associated macroglial and microglial reactions. Knowledge of the distribution of astrocyte cytoplasmic processes is helpful in determining that certain patterns of treatment-related neuropil vacuolation (as well as some artifacts) represent swelling of these processes. On the other hand, vacuoles with different distribution patterns may represent alterations of the myelin sheath. Because brains are typically undersampled for microscopic evaluation, many pathologists are unfamiliar with the circumventricuar organs (CVOs) that represent normal brain structures but are often mistaken for lesions. Therefore, the six CVOs found in the brain are also illustrated in this article.
Collapse
Affiliation(s)
- Robert H. Garman
- Consultants in Veterinary Pathology, Inc., Murrysville, Pennsylvania, USA
| |
Collapse
|
3642
|
Mashanov VS, Zueva OR, Garcia-Arraras JE. Organization of glial cells in the adult sea cucumber central nervous system. Glia 2010; 58:1581-93. [PMID: 20578040 DOI: 10.1002/glia.21031] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The nervous system of echinoderms has long been considered too unique to be directly comparable to the nervous system of other Deuterostomia. Using two novel monoclonal antibodies in combination with epifluorescence, confocal, and electron microscopy, we demonstrate here that the central nervous system of the sea cucumber Holothuria glaberrima possesses a major non-neuronal cell type, which shares striking similarities with the radial glia of chordates. The basic features in common include (a) an elongated shape, (b) long radial processes, (c) short lateral protrusions branching off the main processes and penetrating into the surrounding neuropile, (d) prominent orderly oriented bundles of intermediate filaments, and (e) ability to produce Reissner's substance. Radial glia account for the majority of glia cells in echinoderms and constitutes more than half of the total cell population in the radial nerve cord and about 45% in the circumoral nerve ring. The difference in glia cell number between those regions is significant, suggesting structural specialization within the seemingly simple echinoderm nervous system. Both cell death and proliferation are seen under normal physiological conditions. Although both glia and neurons undergo apoptosis, most of the mitotic cells are identified as radial glia, indicating a key role of this cell type in cell turnover in the nervous system. A hypothesis is proposed that the radial glia could be an ancestral feature of the deuterostome nervous system, and the origin of this cell type might have predated the diversification of the Chordata and Ambulacraria lineages.
Collapse
Affiliation(s)
- Vladimir S Mashanov
- Department of Biology, University of Puerto Rico, Rio Piedras, PR 00936-8377, Puerto Rico.
| | | | | |
Collapse
|
3643
|
Abstract
Reactive astrocytes are a pathological hallmark of many CNS injuries and neurodegenerations. They are characterized by hypertrophy of the soma and processes and an increase in the expression of glial fibrillary acidic protein. Because the cells obscure each other in immunostaining, little is known about the behavior of a single reactive astrocyte, nor how single astrocytes combine to form the glial scar. We have investigated the reaction of fibrous astrocytes to axonal degeneration using a transgenic mouse strain expressing enhanced green fluorescent protein in small subsets of astrocytes. Fibrous astrocytes in the optic nerve and corpus callosum initially react to injury by hypertrophy of the soma and processes. They retract their primary processes, simplifying their shape and dramatically reducing their spatial coverage. At 3 d after crush, quantitative analysis revealed nearly a twofold increase in the thickness of the primary processes, a halving of the number of primary processes leaving the soma and an eightfold reduction in the spatial coverage. In the subsequent week, they partially reextend long processes, returning to a near-normal morphology and an extensive spatial overlap. The resulting glial scar consists of an irregular array of astrocyte processes, contrasting with their original orderly arrangement. These changes are in distinct contrast to those reported for reactive protoplasmic astrocytes of the gray matter, in which the number of processes and branchings increase, but the cells continue to maintain nonoverlapping individual territories throughout their response to injury.
Collapse
|
3644
|
Coracini KF, Fernandes CJ, Barbarini AF, Silva CM, Scabello RT, Oliveira GP, Chadi G. Differential cellular FGF-2 upregulation in the rat facial nucleus following axotomy, functional electrical stimulation and corticosterone: a possible therapeutic target to Bell's palsy. J Brachial Plex Peripher Nerve Inj 2010; 5:16. [PMID: 21062430 PMCID: PMC2995486 DOI: 10.1186/1749-7221-5-16] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2010] [Accepted: 11/09/2010] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND The etiology of Bell's palsy can vary but anterograde axonal degeneration may delay spontaneous functional recovery leading the necessity of therapeutic interventions. Corticotherapy and/or complementary rehabilitation interventions have been employed. Thus the natural history of the disease reports to a neurotrophic resistance of adult facial motoneurons leading a favorable evolution however the related molecular mechanisms that might be therapeutically addressed in the resistant cases are not known. Fibroblast growth factor-2 (FGF-2) pathway signaling is a potential candidate for therapeutic development because its role on wound repair and autocrine/paracrine trophic mechanisms in the lesioned nervous system. METHODS Adult rats received unilateral facial nerve crush, transection with amputation of nerve branches, or sham operation. Other group of unlesioned rats received a daily functional electrical stimulation in the levator labii superioris muscle (1 mA, 30 Hz, square wave) or systemic corticosterone (10 mgkg-1). Animals were sacrificed seven days later. RESULTS Crush and transection lesions promoted no changes in the number of neurons but increased the neurofilament in the neuronal neuropil of axotomized facial nuclei. Axotomy also elevated the number of GFAP astrocytes (143% after crush; 277% after transection) and nuclear FGF-2 (57% after transection) in astrocytes (confirmed by two-color immunoperoxidase) in the ipsilateral facial nucleus. Image analysis reveled that a seven days functional electrical stimulation or corticosterone led to elevations of FGF-2 in the cytoplasm of neurons and in the nucleus of reactive astrocytes, respectively, without astrocytic reaction. CONCLUSION FGF-2 may exert paracrine/autocrine trophic actions in the facial nucleus and may be relevant as a therapeutic target to Bell's palsy.
Collapse
Affiliation(s)
- Karen F Coracini
- Department of Neurology, University of São Paulo, Av. Dr. Arnaldo, 455 2nd floor, room 2119, São Paulo - 01246-903, Brazil
| | - Caio J Fernandes
- Department of Neurology, University of São Paulo, Av. Dr. Arnaldo, 455 2nd floor, room 2119, São Paulo - 01246-903, Brazil
| | - Almir F Barbarini
- Department of Neurology, University of São Paulo, Av. Dr. Arnaldo, 455 2nd floor, room 2119, São Paulo - 01246-903, Brazil
| | - César M Silva
- Department of Neurology, University of São Paulo, Av. Dr. Arnaldo, 455 2nd floor, room 2119, São Paulo - 01246-903, Brazil
| | - Rodrigo T Scabello
- Department of Neurology, University of São Paulo, Av. Dr. Arnaldo, 455 2nd floor, room 2119, São Paulo - 01246-903, Brazil
| | - Gabriela P Oliveira
- Department of Neurology, University of São Paulo, Av. Dr. Arnaldo, 455 2nd floor, room 2119, São Paulo - 01246-903, Brazil
| | - Gerson Chadi
- Department of Neurology, University of São Paulo, Av. Dr. Arnaldo, 455 2nd floor, room 2119, São Paulo - 01246-903, Brazil
| |
Collapse
|
3645
|
Klusa VZ, Isajevs S, Svirina D, Pupure J, Beitnere U, Rumaks J, Svirskis S, Jansone B, Dzirkale Z, Muceniece R, Kalvinsh I, Vinters HV. Neuroprotective properties of mildronate, a small molecule, in a rat model of Parkinson's disease. Int J Mol Sci 2010; 11:4465-87. [PMID: 21151450 PMCID: PMC3000094 DOI: 10.3390/ijms11114465] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2010] [Revised: 10/22/2010] [Accepted: 10/27/2010] [Indexed: 11/16/2022] Open
Abstract
Previously, we have found that mildronate [3-(2,2,2-trimethylhydrazinium) propionate dihydrate], a small molecule with charged nitrogen and oxygen atoms, protects mitochondrial metabolism that is altered by inhibitors of complex I and has neuroprotective effects in an azidothymidine-neurotoxicity mouse model. In the present study, we investigated the effects of mildronate in a rat model of Parkinson's disease (PD) that was generated via a unilateral intrastriatal injection of the neurotoxin 6-hydroxydopamine (6-OHDA). We assessed the expression of cell biomarkers that are involved in signaling cascades and provide neural and glial integration: the neuronal marker TH (tyrosine hydroxylase); ubiquitin (a regulatory peptide involved in the ubiquitin-proteasome degradation system); Notch-3 (a marker of progenitor cells); IBA-1 (a marker of microglial cells); glial fibrillary acidic protein, GFAP (a marker of astrocytes); and inducible nitric oxide synthase, iNOS (a marker of inflammation). The data show that in the 6-OHDA-lesioned striatum, mildronate completely prevented the loss of TH, stimulated Notch-3 expression and decreased the expression of ubiquitin, GFAP and iNOS. These results provide evidence for the ability of mildronate to control the expression of an array of cellular proteins and, thus, impart multi-faceted homeostatic mechanisms in neurons and glial cells in a rat model of PD. We suggest that the use of mildronate provides a protective effect during the early stages of PD that can delay or halt the progression of this neurodegenerative disease.
Collapse
Affiliation(s)
- Vija Z. Klusa
- Department of Pharmacology, Faculty of Medicine, University of Latvia, Riga, Latvia
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +371-292-762-63; Fax: +371-673-663-06
| | - Sergejs Isajevs
- Department of Pathology, Faculty of Medicine, University of Latvia, Riga, Latvia
| | - Darja Svirina
- Department of Pathology, Faculty of Medicine, University of Latvia, Riga, Latvia
| | - Jolanta Pupure
- Department of Pharmacology, Faculty of Medicine, University of Latvia, Riga, Latvia
| | - Ulrika Beitnere
- Department of Pharmacology, Faculty of Medicine, University of Latvia, Riga, Latvia
| | - Juris Rumaks
- Department of Pharmacology, Faculty of Medicine, University of Latvia, Riga, Latvia
| | - Simons Svirskis
- Department of Pharmacology, Faculty of Medicine, University of Latvia, Riga, Latvia
| | - Baiba Jansone
- Department of Pharmacology, Faculty of Medicine, University of Latvia, Riga, Latvia
| | - Zane Dzirkale
- Department of Pharmacology, Faculty of Medicine, University of Latvia, Riga, Latvia
| | - Ruta Muceniece
- Department of Pharmacology, Faculty of Medicine, University of Latvia, Riga, Latvia
| | | | - Harry V. Vinters
- Department of Pathology and Laboratory of Medicine and Neurology, University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
3646
|
Serrano-Pérez MC, Martín ED, Vaquero CF, Azcoitia I, Calvo S, Cano E, Tranque P. Response of transcription factor NFATc3 to excitotoxic and traumatic brain insults: Identification of a subpopulation of reactive astrocytes. Glia 2010; 59:94-107. [DOI: 10.1002/glia.21079] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2010] [Accepted: 08/23/2010] [Indexed: 01/21/2023]
|
3647
|
Brignone MS, Lanciotti A, Macioce P, Macchia G, Gaetani M, Aloisi F, Petrucci TC, Ambrosini E. The beta1 subunit of the Na,K-ATPase pump interacts with megalencephalic leucoencephalopathy with subcortical cysts protein 1 (MLC1) in brain astrocytes: new insights into MLC pathogenesis. Hum Mol Genet 2010; 20:90-103. [PMID: 20926452 DOI: 10.1093/hmg/ddq435] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Megalencephalic leucoencephalopathy with subcortical cysts (MLC) is a rare congenital leucodystrophy caused by mutations in MLC1, a membrane protein of unknown function. MLC1 expression in astrocyte end-feet contacting blood vessels and meninges, along with brain swelling, fluid cysts and myelin vacuolation observed in MLC patients, suggests a possible role for MLC1 in the regulation of fluid and ion homeostasis and cellular volume changes. To identify MLC1 direct interactors and dissect the molecular pathways in which MLC1 is involved, we used NH2-MLC1 domain as a bait to screen a human brain library in a yeast two-hybrid assay. We identified the β1 subunit of the Na,K-ATPase pump as one of the interacting clones and confirmed it by pull-downs, co-fractionation assays and immunofluorescence stainings in human and rat astrocytes in vitro and in brain tissue. By performing ouabain-affinity chromatography on astrocyte and brain extracts, we isolated MLC1 and the whole Na,K-ATPase enzyme in a multiprotein complex that included Kir4.1, syntrophin and dystrobrevin. Because Na,K-ATPase is involved in intracellular osmotic control and volume regulation, we investigated the effect of hypo-osmotic stress on MLC1/Na,K-ATPase relationship in astrocytes. We found that hypo-osmotic conditions increased MLC1 membrane expression and favoured MLC1/Na,K-ATPase-β1 association. Moreover, hypo-osmosis induced astrocyte swelling and the reversible formation of endosome-derived vacuoles, where the two proteins co-localized. These data suggest that through its interaction with Na,K-ATPase, MLC1 is involved in the control of intracellular osmotic conditions and volume regulation in astrocytes, opening new perspectives for understanding the pathological mechanisms of MLC disease.
Collapse
Affiliation(s)
- Maria S Brignone
- Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità, Rome, Italy
| | | | | | | | | | | | | | | |
Collapse
|
3648
|
Lai JCK, Ananthakrishnan G, Jandhyam S, Dukhande VV, Bhushan A, Gokhale M, Daniels CK, Leung SW. Treatment of human astrocytoma U87 cells with silicon dioxide nanoparticles lowers their survival and alters their expression of mitochondrial and cell signaling proteins. Int J Nanomedicine 2010; 5:715-23. [PMID: 21042417 PMCID: PMC2962267 DOI: 10.2147/ijn.s5238] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Recent evidence suggests silicon dioxide micro- and nanoparticles induce cytotoxic effects on lung cells. Thus, there is an increasing concern regarding their potential health hazard. Nevertheless, the putative toxicity of nanoparticles in mammalian cells has not yet been systematically investigated. We previously noted that several metallic oxide nanoparticles exert differential cytotoxic effects on human neural and nonneural cells. Therefore, we hypothesized that silicon dioxide nanoparticles induce cytotoxicity in U87 cells by lowering their survival by decreasing cell survival signaling and disturbing mitochondrial function. To investigate this hypothesis, we determined the activities of the key mitochondrial enzymes, citrate synthase and malate dehydrogenase, in astrocytoma U87 cells treated with silicon dioxide nanoparticles. In addition, we studied the expression of the mitochondrial DNA-encoded proteins, cytochrome C oxidase II and nicotinamide adenine dinucleotide (NADPH) dehydrogenase subunit 6, and cell signaling pathway protein extracellular signal-regulated kinase (ERK) and phosphorylated ERK in treated U87 cells. The activated form of ERK controls cell growth, differentiation, and proliferation. In parallel, we determined survival of U87 cells after treating them with various concentrations of silicon dioxide nanoparticles. Our results indicated that treatment with silicon dioxide nanoparticles induced decreases in U87 cell survival in a dose-related manner. The activities of citrate synthase and malate dehydrogenase in treated U87 cells were increased, possibly due to an energetic compensation in surviving cells. However, the expression of mitochondrial DNA-encoded cytochrome C oxidase subunit II and NADH dehydrogenase subunit 6 and the cell signaling protein ERK and phosphorylated ERK were altered in the treated U87 cells, suggesting that silicon dioxide nanoparticles induced disruption of mitochondrial DNA-encoded protein expression, leading to decreased mitochondrial energy production and decreased cell survival/proliferation signaling. Thus, our results strongly suggest that the cytotoxicity of silicon dioxide nanoparticles in human neural cells implicates altered mitochondrial function and cell survival/proliferation signaling.
Collapse
Affiliation(s)
- James C K Lai
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy and Biomedical Research Institute, Kasiska College of Health Professions
| | | | | | | | | | | | | | | |
Collapse
|
3649
|
Hamby ME, Sofroniew MV. Reactive astrocytes as therapeutic targets for CNS disorders. Neurotherapeutics 2010; 7:494-506. [PMID: 20880511 PMCID: PMC2952540 DOI: 10.1016/j.nurt.2010.07.003] [Citation(s) in RCA: 259] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2010] [Revised: 07/19/2010] [Accepted: 07/20/2010] [Indexed: 12/30/2022] Open
Abstract
Reactive astrogliosis has long been recognized as a ubiquitous feature of CNS pathologies. Although its roles in CNS pathology are only beginning to be defined, genetic tools are enabling molecular dissection of the functions and mechanisms of reactive astrogliosis in vivo. It is now clear that reactive astrogliosis is not simply an all-or-nothing phenomenon but, rather, is a finely gradated continuum of molecular, cellular, and functional changes that range from subtle alterations in gene expression to scar formation. These changes can exert both beneficial and detrimental effects in a context-dependent manner determined by specific molecular signaling cascades. Dysfunction of either astrocytes or the process of reactive astrogliosis is emerging as an important potential source of mechanisms that might contribute to, or play primary roles in, a host of CNS disorders via loss of normal or gain of abnormal astrocyte activities. A rapidly growing understanding of the mechanisms underlying astrocyte signaling and reactive astrogliosis has the potential to open doors to identifying many molecules that might serve as novel therapeutic targets for a wide range of neurological disorders. This review considers general principles and examines selected examples regarding the potential of targeting specific molecular aspects of reactive astrogliosis for therapeutic manipulations, including regulation of glutamate, reactive oxygen species, and cytokines.
Collapse
Affiliation(s)
- Mary E. Hamby
- grid.19006.3e0000000096326718Department of Neurobiology, David Geffen School of Medicine, University of California, 90095 Los Angeles, California
| | - Michael V. Sofroniew
- grid.19006.3e0000000096326718Department of Neurobiology, David Geffen School of Medicine, University of California, 90095 Los Angeles, California
| |
Collapse
|
3650
|
Abstract
Therapeutic management of chronic pain has not been widely successful owing to a lack of understanding of factors that initiate and maintain the chronic pain condition. Efforts to delineate the mechanisms underlying pain long have focused on neuronal elements of pain pathways, and both opiate- and non-opiate-based therapeutics are thought largely to target neurons. Abnormal neuronal activity at the level of spinal cord "pain centers" in the dorsal horn leads to hypersensitivity or a hyperalgesic response subsequent to the initial painful stimulus. Only recently has the experimental literature implicated nonneuronal elements in pain because of the realization that glial-derived signaling molecules can contribute to and modulate pain signaling in the spinal cord. Most notably, glial proinflammatory mediators within the dorsal horn of the spinal cord appear to contribute to self-perpetuating pain. Chronic pain is modeled experimentally through a variety of manipulations of sensory nerves including cutting, crushing, resection, and ligation. The cellular and molecular responses in the spinal cord due to these manipulations often reveal activation of 2 types of glia: microglia and astrocytes. The activation states of both microglia and astrocytes are complex and may be driven by underlying chronic neuropathology and/or a chronically "primed" condition that accounts for their contribution to chronic pain. Recent evidence even suggests that opioid tolerance and withdrawal hyperalgesia may be initiated and maintained via actions of microglia and astroglia. Together, these recent findings suggest that glia will serve as novel therapeutic targets for the treatment of chronic pain. To fully exploit glia as novel therapeutic targets will require a greater understanding of glial biology, as well as the identification of agents able to control the glial reactions involved in chronic pain, without interfering with beneficial glial functions.
Collapse
Affiliation(s)
- James P O'Callaghan
- Centers for Disease Control and Prevention, National Institute for Occupational Safety and Health, Morgantown, WV 26505, USA.
| | | |
Collapse
|