Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 25
1.
Neurochem Res ; 48(4): 1233-1241, 2023 Apr.
Article En | MEDLINE | ID: mdl-36097103

Astrocytes perform a range of homeostatic and regulatory tasks that are critical for normal functioning of the central nervous system. In response to an injury or disease, astrocytes undergo a pronounced transformation into a reactive state that involves changes in the expression of many genes and dramatically changes astrocyte morphology and functions. This astrocyte reactivity is highly dependent on the initiating insult and pathological context. C3a is a peptide generated by the proteolytic cleavage of the third complement component. C3a has been shown to exert neuroprotective effects, stimulate neural plasticity and promote astrocyte survival but can also contribute to synapse loss, Alzheimer's disease type neurodegeneration and blood-brain barrier dysfunction. To test the hypothesis that C3a elicits differential effects on astrocytes depending on their reactivity state, we measured the expression of Gfap, Nes, C3ar1, C3, Ngf, Tnf and Il1b in primary mouse cortical astrocytes after chemical ischemia, after exposure to lipopolysaccharide (LPS) as well as in control naïve astrocytes. We found that C3a down-regulated the expression of Gfap, C3 and Nes in astrocytes after ischemia. Further, C3a increased the expression of Tnf and Il1b in naive astrocytes and the expression of Nes in astrocytes exposed to LPS but did not affect the expression of C3ar1 or Ngf. Jointly, these results provide the first evidence that the complement peptide C3a modulates the responses of astrocytes in a highly context-dependent manner.


Astrocytes , Lipopolysaccharides , Mice , Animals , Astrocytes/metabolism , Lipopolysaccharides/pharmacology , Blood-Brain Barrier/metabolism , Complement C3a/metabolism , Peptides/metabolism
2.
Prog Neurobiol ; 209: 102199, 2022 02.
Article En | MEDLINE | ID: mdl-34921928

Restoration of functional connectivity is a major contributor to functional recovery after stroke. We investigated the role of reactive astrocytes in functional connectivity and recovery after photothrombotic stroke in mice with attenuated reactive gliosis (GFAP-/-Vim-/-). Infarct volume and longitudinal functional connectivity changes were determined by in vivo T2-weighted magnetic resonance imaging (MRI) and resting-state functional MRI. Sensorimotor function was assessed with behavioral tests, and glial and neural plasticity responses were quantified in the peri-infarct region. Four weeks after stroke, GFAP-/-Vim-/- mice showed impaired recovery of sensorimotor function and aberrant restoration of global neuronal connectivity. These mice also exhibited maladaptive plasticity responses, shown by higher number of lost and newly formed functional connections between primary and secondary targets of cortical stroke regions and increased peri-infarct expression of the axonal plasticity marker Gap43. We conclude that reactive astrocytes modulate recovery-promoting plasticity responses after ischemic stroke.


Ischemic Stroke , Stroke , Animals , Astrocytes/metabolism , Gliosis/metabolism , Humans , Mice , Neuronal Plasticity , Recovery of Function/physiology
3.
Acta Physiol (Oxf) ; 228(3): e13399, 2020 03.
Article En | MEDLINE | ID: mdl-31597221

AIM: Astrocytes play a homeostatic role in the central nervous system and influence numerous aspects of neurophysiology via intracellular trafficking of vesicles. Intermediate filaments (IFs), also known as nanofilaments, regulate a number of cellular processes including organelle trafficking and adult hippocampal neurogenesis. We have recently demonstrated that the IF protein nestin, a marker of neural stem cells and immature and reactive astrocytes, is also expressed in some astrocytes in the unchallenged hippocampus and regulates neurogenesis through Notch signalling from astrocytes to neural stem cells, possibly via altered trafficking of vesicles containing the Notch ligand Jagged-1. METHODS: We thus investigated whether nestin affects vesicle dynamics in astrocytes by examining single vesicle interactions with the plasmalemma and vesicle trafficking with high-resolution cell-attached membrane capacitance measurements and confocal microscopy. We used cell cultures of astrocytes from nestin-deficient (Nes-/- ) and wild-type (wt) mice, and fluorescent dextran and Fluo-2 to examine vesicle mobility and intracellular Ca2+ concentration respectively. RESULTS: Nes-/- astrocytes exhibited altered sizes of vesicles undergoing full fission and transient fusion, altered vesicle fusion pore geometry and kinetics, decreased spontaneous vesicle mobility and altered ATP-evoked mobility. Purinergic stimulation evoked Ca2+ signalling that was slightly attenuated in Nes-/- astrocytes, which exhibited more oscillatory Ca2+ responses than wt astrocytes. CONCLUSION: These results demonstrate at the single vesicle level that nestin regulates vesicle interactions with the plasmalemma and vesicle trafficking, indicating its potential role in astrocyte vesicle-based communication.


Adenosine Triphosphate/metabolism , Astrocytes/metabolism , Calcium/metabolism , Cell Membrane/metabolism , Nestin/metabolism , Animals , Biological Transport , Cell Fusion , Cells, Cultured , Exocytosis/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nestin/genetics , Signal Transduction
4.
Cells ; 8(9)2019 09 01.
Article En | MEDLINE | ID: mdl-31480524

Vimentin (VIM) is an intermediate filament (nanofilament) protein expressed in multiple cell types, including astrocytes. Mice with VIM mutations of serine sites phosphorylated during mitosis (VIMSA/SA) show cytokinetic failure in fibroblasts and lens epithelial cells, chromosomal instability, facilitated cell senescence, and increased neuronal differentiation of neural progenitor cells. Here we report that in vitro immature VIMSA/SA astrocytes exhibit cytokinetic failure and contain vimentin accumulations that co-localize with mitochondria. This phenotype is transient and disappears with VIMSA/SA astrocyte maturation and expression of glial fibrillary acidic protein (GFAP); it is also alleviated by the inhibition of cell proliferation. To test the hypothesis that GFAP compensates for the effect of VIMSA/SA in astrocytes, we crossed the VIMSA/SA and GFAP-/- mice. Surprisingly, the fraction of VIMSA/SA immature astrocytes with abundant vimentin accumulations was reduced when on GFAP-/- background. This indicates that the disappearance of vimentin accumulations and cytokinetic failure in mature astrocyte cultures are independent of GFAP expression. Both VIMSA/SA and VIMSA/SAGFAP-/- astrocytes showed normal mitochondrial membrane potential and vulnerability to H2O2, oxygen/glucose deprivation, and chemical ischemia. Thus, mutation of mitotic phosphorylation sites in vimentin triggers formation of vimentin accumulations and cytokinetic failure in immature astrocytes without altering their vulnerability to oxidative stress.


Astrocytes/metabolism , Cell Division , Neurogenesis , Vimentin/metabolism , Animals , Astrocytes/cytology , Astrocytes/physiology , Cells, Cultured , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Mice , Mice, Inbred C57BL , Mutation , Phosphorylation , Protein Domains , Vimentin/chemistry , Vimentin/genetics
5.
Biol Chem ; 400(9): 1147-1156, 2019 08 27.
Article En | MEDLINE | ID: mdl-31063456

Intermediate filaments (also termed nanofilaments) are involved in many cellular functions and play important roles in cellular responses to stress. The upregulation of glial fibrillary acidic protein (GFAP) and vimentin (Vim), intermediate filament proteins of astrocytes, is the hallmark of astrocyte activation and reactive gliosis in response to injury, ischemia or neurodegeneration. Reactive gliosis is essential for the protective role of astrocytes at acute stages of neurotrauma or ischemic stroke. However, GFAP and Vim were also linked to neural plasticity and regenerative responses in healthy and injured brain. Mice deficient for GFAP and vimentin (GFAP-/-Vim-/-) exhibit increased post-traumatic synaptic plasticity and increased basal and post-traumatic hippocampal neurogenesis. Here we assessed the locomotor and exploratory behavior of GFAP-/-Vim-/- mice, their learning, memory and memory extinction, by using the open field, object recognition and Morris water maze tests, trace fear conditioning, and by recording reversal learning in IntelliCages. While the locomotion, exploratory behavior and learning of GFAP-/-Vim-/- mice, as assessed by object recognition, the Morris water maze, and trace fear conditioning tests, were comparable to wildtype mice, GFAP-/-Vim-/- mice showed more pronounced memory extinction when tested in IntelliCages, a finding compatible with the scenario of an increased rate of reorganization of the hippocampal circuitry.


Glial Fibrillary Acidic Protein/physiology , Learning/physiology , Memory/physiology , Vimentin/physiology , Animals , Glial Fibrillary Acidic Protein/genetics , Hippocampus/physiology , Intermediate Filaments/metabolism , Male , Maze Learning , Mice , Mice, Knockout , Neurogenesis , Vimentin/genetics
6.
Free Radic Biol Med ; 120: 380-394, 2018 05 20.
Article En | MEDLINE | ID: mdl-29635011

The type III intermediate filament protein glial fibrillary acidic protein (GFAP) contributes to the homeostasis of astrocytes, where it co-polymerizes with vimentin. Conversely, alterations in GFAP assembly or degradation cause intracellular aggregates linked to astrocyte dysfunction and neurological disease. Moreover, injury and inflammation elicit extensive GFAP organization and expression changes, which underline reactive gliosis. Here we have studied GFAP as a target for modification by electrophilic inflammatory mediators. We show that the GFAP cysteine, C294, is targeted by lipoxidation by cyclopentenone prostaglandins (cyPG) in vitro and in cells. Electrophilic modification of GFAP in cells leads to a striking filament rearrangement, with retraction from the cell periphery and juxtanuclear condensation in thick bundles. Importantly, the C294S mutant is resistant to cyPG addition and filament disruption, thus highlighting the critical role of this residue as a sensor of oxidative damage. However, GFAP C294S shows defective or delayed network formation in GFAP-deficient cells, including SW13/cl.2 cells and GFAP- and vimentin-deficient primary astrocytes. Moreover, GFAP C294S does not effectively integrate with and even disrupts vimentin filaments in the short-term. Interestingly, short-spacer bifunctional cysteine crosslinking produces GFAP-vimentin heterodimers, suggesting that a certain proportion of cysteine residues from both proteins are spatially close. Collectively, these results support that the conserved cysteine residue in type III intermediate filament proteins serves as an electrophilic stress sensor and structural element. Therefore, oxidative modifications of this cysteine could contribute to GFAP disruption or aggregation in pathological situations associated with oxidative or electrophilic stress.


Astrocytes/chemistry , Astrocytes/metabolism , Cysteine/chemistry , Glial Fibrillary Acidic Protein/chemistry , Glial Fibrillary Acidic Protein/metabolism , Animals , Cattle , Cysteine/metabolism , Humans , Intermediate Filament Proteins/chemistry , Intermediate Filament Proteins/metabolism , Mice , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Oxidation-Reduction , Oxidative Stress/physiology , Prostaglandins/chemistry , Prostaglandins/metabolism
7.
Brain Res Bull ; 136: 130-138, 2018 01.
Article En | MEDLINE | ID: mdl-28159699

In response to central nervous system (CNS) injury, astrocytes upregulate intermediate filament (nanofilament) proteins GFAP and vimentin. Whereas the intermediate filament upregulation in astrocytes is important for neuroprotection in the acute phase of injury, in some contexts it might inhibit some of the regenerative processes later on. Thus, timely modulation of the astrocyte intermediate filaments was proposed as a strategy to promote brain repair. We used clomipramine, epoxomicin and withaferin A, drugs reported to decrease the expression of GFAP, and assessed their effect on neurosupportive properties and resilience of astrocytes to oxygen and glucose deprivation (OGD). Clomipramine decreased protein levels of GFAP, as well as vimentin and nestin, and did not affect astrocyte resilience to oxidative stress. Withaferin A sensitized astrocytes to OGD. Both clomipramine and epoxomicin promoted the attachment and survival of neurons co-cultured with astrocytes under standard culture conditions. Moreover, epoxomicin increased neurosupportive properties of astrocytes after OGD. Our data point to clomipramine and epoxomicin as potential candidates for astrocyte modulation to improve outcome after CNS injury.


Astrocytes/drug effects , Clomipramine/pharmacology , Intermediate Filaments/drug effects , Neuroprotective Agents/pharmacology , Withanolides/pharmacology , Animals , Astrocytes/metabolism , Brain/drug effects , Brain/metabolism , Cell Hypoxia/drug effects , Cell Hypoxia/physiology , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Clomipramine/toxicity , Coculture Techniques , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Glucose/deficiency , Intermediate Filaments/genetics , Intermediate Filaments/metabolism , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Neurons/drug effects , Neurons/physiology , Neuroprotective Agents/toxicity , Oligopeptides/pharmacology , Oligopeptides/toxicity , Reactive Oxygen Species/metabolism , Withanolides/toxicity
8.
Cereb Cortex ; 27(6): 3360-3377, 2017 06 01.
Article En | MEDLINE | ID: mdl-28398520

Microglia and astrocytes have been considered until now as cells with very distinct identities. Here, we assessed the heterogeneity within microglia/monocyte cell population in mouse hippocampus and determined their response to injury, by using single-cell gene expression profiling of cells isolated from uninjured and deafferented hippocampus. We found that in individual cells, microglial markers Cx3cr1, Aif1, Itgam, and Cd68 were co-expressed. Interestingly, injury led to the co-expression of the astrocyte marker Gfap in a subpopulation of Cx3cr1-expressing cells from both the injured and contralesional hippocampus. Cells co-expressing astrocyte and microglia markers were also detected in the in vitro LPS activation/injury model and in sections from human brain affected by stroke, Alzheimer's disease, and Lewy body dementia. Our findings indicate that injury and chronic neurodegeneration lead to the appearance of cells that share molecular characteristics of both microglia and astrocytes, 2 cell types with distinct embryologic origin and function.


Astrocytes/pathology , Brain Injuries/pathology , Entorhinal Cortex/pathology , Gene Expression Regulation/physiology , Microglia/pathology , Alzheimer Disease/pathology , Animals , Apoptosis Inducing Factor/genetics , Apoptosis Inducing Factor/metabolism , Astrocytes/metabolism , Brain Injuries/metabolism , CD11b Antigen/genetics , CD11b Antigen/metabolism , CX3C Chemokine Receptor 1/genetics , CX3C Chemokine Receptor 1/metabolism , Cell Hypoxia/drug effects , Cells, Cultured , Dementia/pathology , Glial Fibrillary Acidic Protein/metabolism , Glucose/deficiency , Hippocampus/pathology , Humans , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/drug effects , Microglia/metabolism
9.
Mol Neurobiol ; 53(5): 3076-3087, 2016 07.
Article En | MEDLINE | ID: mdl-25972241

Astrocytes are the most numerous cells in the central nervous system with a range of homeostatic and regulatory functions. Under normal conditions as well as after ischemia, astrocytes promote neuronal survival. We have previously reported that the complement-derived peptide C3a stimulates neuronal differentiation of neural progenitor cells and protects the immature brain tissue against hypoxic-ischemic injury. Here, we studied the effects of C3a on the response of mouse cortical astrocytes to ischemia. We have found that chemical ischemia, induced by combined inhibition of oxidative phosphorylation and glycolysis, upregulates the expression of C3a receptor in cultured primary astrocytes. C3a treatment protected wild-type but not C3a receptor-deficient astrocytes from cell death induced by chemical ischemia or oxygen-glucose deprivation by reducing ERK signaling and caspase-3 activation. C3a attenuated ischemia-induced upregulation of glial fibrillary acidic protein; however, the protective effects of C3a were not dependent on the presence of the astrocyte intermediate filament system. Pre-treatment of astrocytes with C3a during recovery abrogated the ischemia-induced neuroprotective phenotype of astrocytes. Jointly, these results provide the first evidence that the complement peptide C3a modulates the response of astrocytes to ischemia and increases their ability to cope with ischemic stress.


Astrocytes/enzymology , Brain Ischemia/drug therapy , Brain Ischemia/pathology , Complement C3a/therapeutic use , Stress, Physiological , Animals , Astrocytes/drug effects , Astrocytes/pathology , Caspase 3/metabolism , Cell Survival/drug effects , Cerebral Cortex/pathology , Coculture Techniques , Complement C3a/pharmacology , Enzyme Activation/drug effects , Intermediate Filaments/metabolism , MAP Kinase Signaling System/drug effects , Mice, Inbred C57BL , Nerve Growth Factor/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Complement/metabolism , Stress, Physiological/drug effects , Up-Regulation/drug effects
10.
Tissue Eng Part C Methods ; 20(6): 485-92, 2014 Jun.
Article En | MEDLINE | ID: mdl-24102451

Neuronal signal transduction and communication in vivo is based on highly complex and dynamic networks among neurons expanding in a three-dimensional (3D) manner. Studies of cell-cell communication, synaptogenesis, and neural network plasticity constitute major research areas for understanding the involvement of neurons in neurodegenerative diseases, such as Huntington's, Alzheimer's, and Parkinson's disease, and in regenerative neural plasticity responses in situations, such as neurotrauma or stroke. Various cell culture systems constitute important experimental platforms to study neuronal functions in health and disease. A major downside of the existing cell culture systems is that the alienating planar cell environment leads to aberrant cell-cell contacts and network formation and increased reactivity of cell culture-contaminating glial cells. To mimic a suitable 3D environment for the growth and investigation of neuronal networks in vitro has posed an insurmountable challenge. Here, we report the development of a novel electrospun, polyurethane nanofiber-based 3D cell culture system for the in vitro support of neuronal networks, in which neurons can grow freely in all directions and form network structures more complex than any culture system has so far been able to support. In this 3D system, neurons extend processes from their cell bodies as a function of the nanofiber diameter. The nanofiber scaffold also minimizes the reactive state of contaminating glial cells.


Hippocampus/cytology , Nanofibers/chemistry , Nerve Net/physiology , Neurons/cytology , Neurons/physiology , Printing, Three-Dimensional , Tissue Scaffolds , Animals , Batch Cell Culture Techniques/instrumentation , Cell Adhesion/physiology , Cell Proliferation/physiology , Cell Survival/physiology , Cells, Cultured , Electroplating/methods , Hippocampus/physiology , Humans , Mice , Mice, Inbred C57BL , Nanofibers/ultrastructure , Nerve Net/cytology , Particle Size , Rotation , Tissue Engineering/instrumentation , Tissue Engineering/methods
11.
J Neurochem ; 128(6): 878-89, 2014 Mar.
Article En | MEDLINE | ID: mdl-24188029

Heparin-binding epidermal growth factor-like growth factor (HB-EGF), a vascular-derived trophic factor, belongs to the epidermal growth factor (EGF) family of neuroprotective, hypoxia-inducible proteins released by astrocytes in CNS injuries. It was suggested that HB-EGF can replace fetal calf serum (FCS) in astrocyte cultures. We previously demonstrated that in contrast to standard 2D cell culture systems, Bioactive3D culture system, when used with FCS, minimizes the baseline activation of astrocytes and preserves their complex morphology. Here, we show that HB-EGF induced EGF receptor (EGFR) activation by Y1068 phosphorylation, Mapk/Erk pathway activation, and led to an increase in cell proliferation, more prominent in Bioactive3D than in 2D cultures. HB-EGF changed morphology of 2D and Bioactive3D cultured astrocytes toward a radial glia-like phenotype and induced the expression of intermediate filament and progenitor cell marker protein nestin. Glial fibrillary acidic protein (GFAP) and vimentin protein expression was unaffected. RT-qPCR analysis demonstrated that HB-EGF affected the expression of Notch signaling pathway genes, implying a role for the Notch signaling in HB-EGF-mediated astrocyte response. HB-EGF can be used as a FCS replacement for astrocyte expansion and in vitro experimentation both in 2D and Bioactive3D culture systems; however, caution should be exercised since it appears to induce partial de-differentiation of astrocytes.


Astrocytes/cytology , Astrocytes/metabolism , Intercellular Signaling Peptides and Proteins/physiology , Intermediate Filament Proteins/metabolism , MAP Kinase Signaling System/physiology , Animals , Astrocytes/drug effects , Cell Culture Techniques/methods , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Proliferation/drug effects , Cells, Cultured , Female , Glial Fibrillary Acidic Protein , Heparin-binding EGF-like Growth Factor , Intercellular Signaling Peptides and Proteins/pharmacology , MAP Kinase Signaling System/drug effects , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Nerve Tissue Proteins/metabolism , Nestin/metabolism , Receptors, Notch/metabolism , Vimentin/metabolism
12.
Histochem Cell Biol ; 140(1): 81-91, 2013 Jul.
Article En | MEDLINE | ID: mdl-23756782

As a response to central nervous system injury, astrocytes become reactive. Two cellular hallmarks of reactive gliosis are hypertrophy of astrocyte processes and upregulation of intermediate filament (nanofilament) proteins glial fibrillary acidic protein (GFAP), vimentin, nestin, and synemin. Astrocytes in mice devoid of GFAP and vimentin (GFAP (-/-) Vim (-/-)) do not form cytoplasmic intermediate filaments. GFAP (-/-) Vim (-/-) mice develop larger infarcts after ischemic stroke (Li et al. in J Cereb Blood Flow Metab 28(3):468-481, 2008). Here, we attempted to analyze the underlying mechanisms using oxygen-glucose deprivation (OGD), an in vitro ischemia model, examining a potential link between astrocyte intermediate filaments and reactive oxygen species (ROS). We observed a reorganization of the intermediate filament network in astrocytes exposed to OGD. ROS accumulation was higher in GFAP (-/-) Vim (-/-) than wild-type astrocytes when exposed to OGD followed by reperfusion or when exposed to hydrogen peroxide. These results indicate that the elimination of ROS is impaired in the absence of the intermediate filament system. Compared to wild-type astrocytes, GFAP (-/-) Vim (-/-) astrocytes exposed to OGD and reperfusion exhibited increased cell death and conferred lower degree of protection to cocultured neurons. We conclude that the astrocyte intermediate filament system is important for the cell response to oxidative stress induced by OGD followed by reperfusion.


Astrocytes/pathology , Glucose/metabolism , Intermediate Filaments/pathology , Oxidative Stress/physiology , Oxygen/metabolism , Reactive Oxygen Species/analysis , Animals , Cell Survival , Cells, Cultured , Mice , Mice, Knockout , Mutation , Reperfusion
13.
Glia ; 61(3): 432-40, 2013 Mar.
Article En | MEDLINE | ID: mdl-23292921

We tested the hypothesis that astrocytes grown in a suitable three-dimensional (3D) cell culture system exhibit morphological and biochemical features of in vivo astrocytes that are otherwise lost upon transfer from the in vivo to a two-dimensional (2D) culture environment. First, we report development of a novel bioactively coated nanofiber-based 3D culture system (Bioactive3D) that supports cultures of primary mouse astrocytes. Second, we show that Bioactive3D culture system maintains the in vivo-like morphological complexity of cultured cells, allows movement of astrocyte filopodia in a way that resembles the in vivo situation, and also minimizes the cellular stress, an inherent feature of standard 2D cell culture systems. Third, we demonstrate that the expression of gap junctions is reduced in astrocytes cultured in a 3D system that supports well-organized cell-cell communication, in contrast to the enforced planar tiling of cells in a standard 2D system. Finally, we show that astrocytes cultured in the Bioactive3D system do not show the undesired baseline activation but are fully responsive to activation-inducing stimuli. Thus, astrocytes cultured in the Bioactive3D appear to more closely resemble astrocytes in vivo and represent a superior in vitro system for assessing (patho)physiological and pharmacological responses of these cells and potentially also in co-cultures of astrocytes and other cell types.


Astrocytes/cytology , Brain/cytology , Cell Culture Techniques/methods , Animals , Cell Shape , Mice
14.
Stem Cells ; 30(10): 2320-9, 2012 Oct.
Article En | MEDLINE | ID: mdl-22887872

Adult neurogenesis is regulated by a number of cellular players within the neurogenic niche. Astrocytes participate actively in brain development, regulation of the mature central nervous system (CNS), and brain plasticity. They are important regulators of the local environment in adult neurogenic niches through the secretion of diffusible morphogenic factors, such as Wnts. Astrocytes control the neurogenic niche also through membrane-associated factors, however, the identity of these factors and the mechanisms involved are largely unknown. In this study, we sought to determine the mechanisms underlying our earlier finding of increased neuronal differentiation of neural progenitor cells when cocultured with astrocytes lacking glial fibrillary acidic protein (GFAP) and vimentin (GFAP(-/-) Vim(-/-) ). We used primary astrocyte and neurosphere cocultures to demonstrate that astrocytes inhibit neuronal differentiation through a cell-cell contact. GFAP(-/-) Vim(-/-) astrocytes showed reduced endocytosis of Notch ligand Jagged1, reduced Notch signaling, and increased neuronal differentiation of neurosphere cultures. This effect of GFAP(-/-) Vim(-/-) astrocytes was abrogated in the presence of immobilized Jagged1 in a manner dependent on the activity of γ-secretase. Finally, we used GFAP(-/-) Vim(-/-) mice to show that in the absence of GFAP and vimentin, hippocampal neurogenesis under basal conditions as well as after injury is increased. We conclude that astrocytes negatively regulate neurogenesis through the Notch pathway, and endocytosis of Notch ligand Jagged1 in astrocytes and Notch signaling from astrocytes to neural stem/progenitor cells depends on the intermediate filament proteins GFAP and vimentin.


Astrocytes/metabolism , Calcium-Binding Proteins/genetics , Intercellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , Neurogenesis/genetics , Receptors, Notch/genetics , Vimentin/genetics , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Animals , Astrocytes/cytology , Calcium-Binding Proteins/metabolism , Cell Communication/genetics , Cell Differentiation , Coculture Techniques , Endocytosis , Gene Expression Regulation, Developmental , Glial Fibrillary Acidic Protein , Intercellular Signaling Peptides and Proteins/metabolism , Jagged-1 Protein , Male , Membrane Proteins/metabolism , Mice , Mice, Knockout , Nerve Tissue Proteins/deficiency , Primary Cell Culture , Receptors, Notch/metabolism , Serrate-Jagged Proteins , Signal Transduction , Stem Cells/cytology , Stem Cells/metabolism , Vimentin/deficiency , Wnt Proteins/genetics , Wnt Proteins/metabolism
15.
J Neuroinflammation ; 9: 144, 2012 Jun 26.
Article En | MEDLINE | ID: mdl-22734718

BACKGROUND: In immune-mediated diseases of the central nervous system, astrocytes exposed to interferon-γ (IFN-γ) can express major histocompatibility complex (MHC) class II molecules and antigens on their surface. MHC class II molecules are thought to be delivered to the cell surface by membrane-bound vesicles. However, the characteristics and dynamics of this vesicular traffic are unclear, particularly in reactive astrocytes, which overexpress intermediate filament (IF) proteins that may affect trafficking. The aim of this study was to determine the mobility of MHC class II vesicles in wild-type (WT) astrocytes and in astrocytes devoid of IFs. METHODS: The identity of MHC class II compartments in WT and IF-deficient astrocytes 48 h after IFN-γ activation was determined immunocytochemically by using confocal microscopy. Time-lapse confocal imaging and Alexa Fluor546-dextran labeling of late endosomes/lysosomes in IFN-γ treated cells was used to characterize the motion of MHC class II vesicles. The mobility of vesicles was analyzed using ParticleTR software. RESULTS: Confocal imaging of primary cultures of WT and IF-deficient astrocytes revealed IFN-γ induced MHC class II expression in late endosomes/lysosomes, which were specifically labeled with Alexa Fluor546-conjugated dextran. Live imaging revealed faster movement of dextran-positive vesicles in IFN-γ-treated than in untreated astrocytes. Vesicle mobility was lower in IFN-γ-treated IF-deficient astrocytes than in WT astrocytes. Thus, the IFN-γ-induced increase in the mobility of MHC class II compartments is IF-dependent. CONCLUSIONS: Since reactivity of astrocytes is a hallmark of many CNS pathologies, it is likely that the up-regulation of IFs under such conditions allows a faster and therefore a more efficient delivery of MHC class II molecules to the cell surface. In vivo, such regulatory mechanisms may enable antigen-presenting reactive astrocytes to respond rapidly and in a controlled manner to CNS inflammation.


Astrocytes/metabolism , Cell Compartmentation/physiology , Histocompatibility Antigens Class II/metabolism , Interferon-gamma/physiology , Intermediate Filament Proteins/physiology , Animals , Cells, Cultured , Interferon-gamma/genetics , Intermediate Filament Proteins/genetics , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Protein Transport/genetics , Protein Transport/physiology , Up-Regulation/genetics , Up-Regulation/physiology
16.
J Cell Sci ; 124(Pt 16): 2711-22, 2011 Aug 15.
Article En | MEDLINE | ID: mdl-21807936

Aurora A kinase is overexpressed in the majority of breast carcinomas. A chemical genetic approach was used to identify the malignant targets of Aurora A, which revealed pleckstrin-homology-like domain protein PHLDA1 as an Aurora A substrate. PHLDA1 downregulation is a powerful prognostic predictor for breast carcinoma, which was confirmed in our study. We further show that downregulation of PHLDA1 is associated with estrogen receptor (ER) expression in breast carcinoma. Aurora A directly phosphorylates PHLDA1 leading to its degradation. PHLDA1 also negatively regulates Aurora A, thereby triggering a feedback loop. We demonstrate the underlying mechanisms by which PHLDA1 upregulation strongly antagonizes Aurora-A-mediated oncogenic pathways, thereby revealing PHLDA1 degradation as a key mechanism by which Aurora A promotes breast malignancy. Thus, not surprisingly, PHLDA1 upregulation acts synergistically with Aurora A inhibition in promoting cell death. PHLDA1 overexpression might therefore be an alternative method to modulate Aurora A deregulation in breast carcinoma. Finally, this study led to the discovery of a mutation in the Aurora A active site that renders it amenable to the chemical genetic approach. Similar mutations are required for Aurora B, suggesting that this modified approach can be extended to other kinases that have hitherto not been amenable to this methodology.


Breast Neoplasms/metabolism , Carcinoma/metabolism , Feedback, Physiological , Protein Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism , Apoptosis/genetics , Aurora Kinase B , Aurora Kinases , Biomarkers, Tumor/metabolism , Breast Neoplasms/diagnosis , Breast Neoplasms/pathology , Carcinoma/diagnosis , Carcinoma/pathology , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Humans , Mutagenesis, Site-Directed , Mutation/genetics , Protein Engineering , Protein Serine-Threonine Kinases/genetics , RNA, Small Interfering/genetics , Receptors, Estrogen/metabolism , Substrate Specificity , Transcription Factors/genetics , Transgenes/genetics
17.
Mol Biol Cell ; 22(9): 1452-62, 2011 May.
Article En | MEDLINE | ID: mdl-21389115

Nuclear fragmentation is a common feature in many neurodegenerative diseases, including Alzheimer's disease (AD). In this study, we show that nuclear lamina dispersion is an early and irreversible trigger for cell death initiated by deregulated Cdk5, rather than a consequence of apoptosis. Cyclin-dependent kinase 5 (Cdk5) activity is significantly increased in AD and contributes to all three hallmarks: neurotoxic amyloid-ß (Aß), neurofibrillary tangles (NFT), and extensive cell death. Using Aß and glutamate as the neurotoxic stimuli, we show that deregulated Cdk5 induces nuclear lamina dispersion by direct phosphorylation of lamin A and lamin B1 in neuronal cells and primary cortical neurons. Phosphorylation-resistant mutants of lamins confer resistance to nuclear dispersion and cell death on neurotoxic stimulation, highlighting this as a major mechanism for neuronal death. Rapid alteration of lamin localization pattern and nuclear membrane change are further supported by in vivo data using an AD mouse model. After p25 induction, the pattern of lamin localization was significantly altered, preceding neuronal death, suggesting that it is an early pathological event in p25-inducible transgenic mice. Importantly, lamin dispersion is coupled with Cdk5 nuclear localization, which is highly neurotoxic. Inhibition of nuclear dispersion rescues neuronal cells from cell death, underscoring the significance of this event to Cdk5-mediated neurotoxicity.


Cyclin-Dependent Kinase 5/metabolism , Neurons/pathology , Nuclear Envelope/enzymology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/pharmacology , Animals , Cell Death , Cyclin-Dependent Kinase 5/genetics , Disease Models, Animal , Glutamic Acid/pharmacology , JNK Mitogen-Activated Protein Kinases/metabolism , Lamin Type A/chemistry , Lamin Type A/genetics , Lamin Type A/metabolism , Lamin Type B/chemistry , Lamin Type B/genetics , Lamin Type B/metabolism , Mice , Mice, Transgenic , Mutation , Nerve Tissue Proteins/metabolism , Neurofibrillary Tangles , Neurons/metabolism , Nuclear Lamina/pathology , Phosphorylation , Phosphotransferases , Rats , Rats, Sprague-Dawley
18.
J Neurochem ; 117(3): 479-93, 2011 May.
Article En | MEDLINE | ID: mdl-21332718

The nerve growth factor receptor TrkA (tropomyosin-related kinase receptor) participates in the survival and differentiation of several neuronal populations. The C-terminal tail of TrkA contains a PPXY motif, the binding site of the E3 ubiquitin-ligase Nedd4-2 (neural precursor cell expressed, developmentally down-regulated 4-2). In order to analyze the role of Nedd4-2 ubiquitination on TrkA function, we generated three TrkA mutants, by introducing point mutations on conserved hydrophobic amino acids - Leu784 and Val790 switched to Ala. TrkA mutants co-localized and co-immunoprecipitated more efficiently with Nedd4-2 and consequently a strong increase in the basal multimonoubiquitination of the mutant receptors was observed. In addition, we found a decrease in TrkA abundance because of the preferential sorting of mutant receptors towards the late endosome/lysosome pathway instead of recycling back to the plasma membrane. Despite the reduction in the amount of membrane receptor caused by the C-terminal changes, TrkA mutants were able to activate signaling cascades and were even more efficient in promoting neurite outgrowth than the wild-type receptor. Our results demonstrate that the C-terminal tail hydrophobicity of TrkA regulates Nedd4-2 binding and activity and therefore controls receptor turnover. In addition, TrkA multimonoubiquitination does not interfere with the activation of signaling cascades, but rather potentiates receptor signaling leading to differentiation.


Cysteine Endopeptidases/metabolism , Lysosomes/metabolism , Receptor, trkA/metabolism , Signal Transduction/physiology , Ubiquitination/physiology , Animals , Biotin/metabolism , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cysteine Endopeptidases/genetics , Down-Regulation/drug effects , Down-Regulation/genetics , Green Fluorescent Proteins/genetics , Immunoprecipitation/methods , Lysosomal Membrane Proteins/genetics , Lysosomal Membrane Proteins/metabolism , Lysosomes/drug effects , Mutation/genetics , Nerve Growth Factor/pharmacology , PC12 Cells , Protein Binding/genetics , Protein Interaction Domains and Motifs/genetics , Protein Transport/drug effects , Rats , Receptor, trkA/genetics , Signal Transduction/drug effects , Transfection/methods , Ubiquitination/drug effects , rab4 GTP-Binding Proteins/genetics , rab4 GTP-Binding Proteins/metabolism , rab5 GTP-Binding Proteins/genetics , rab5 GTP-Binding Proteins/metabolism
19.
J Neurochem ; 113(5): 1221-9, 2010 Jun.
Article En | MEDLINE | ID: mdl-20345761

Cyclin-dependent kinase (Cdk) 5 and p38 activities are significantly increased in Alzheimer's Disease (AD). Both p38 and Cdk5 promote neurodegeneration upon deregulation. However, to date the mechanistic link between Cdk5 and p38 remains unclear. This study presents the first mechanism showing Cdk5 as a major regulator of p38 cascade in neurons and in transgenic mouse model of AD. Using beta-amyloid and glutamate as the neurotoxic stimuli, our results show that deregulated Cdk5 induces p38 activation by increasing reactive oxygen species (ROS) in neuronal cells and in primary cortical neurons. Elimination of ROS inhibits p38 activation, revealing ROS as major stimuli of the p38 cascade. Importantly, Cdk5-mediated p38 activation increases c-Jun expression, thereby revealing a mechanistic link between deregulated Cdk5 and c-Jun level in AD brains. c-Jun is over-expressed in AD, and is believed to contribute significantly to neurodegeneration. Based on the proposed mechanism, Cdk5 inhibition is more neuroprotective relative to p38 and c-Jun, suggesting that Cdk5 is an upstream regulator of neurodegenerative pathways triggered by p38 and a preferable therapeutic target for AD.


Alzheimer Disease/pathology , Cyclin-Dependent Kinase 5/physiology , Neurons/pathology , p38 Mitogen-Activated Protein Kinases/physiology , Alzheimer Disease/enzymology , Amyloid beta-Peptides/pharmacology , Animals , Blotting, Western , Calpain/physiology , Coloring Agents , Cyclin-Dependent Kinase 5/antagonists & inhibitors , Glutamic Acid/pharmacology , Humans , Immunohistochemistry , MAP Kinase Kinase 6/metabolism , Mice , Mice, Transgenic , Neurons/enzymology , Phosphorylation , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Signal Transduction/physiology , Tetrazolium Salts , Thiazoles , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
20.
J Neurochem ; 107(1): 265-78, 2008 Oct.
Article En | MEDLINE | ID: mdl-18691386

Oxidative stress is one of the earliest events in Alzheimer's disease (AD). A chemical genetic screen revealed that deregulated cyclin-dependent kinase 5 (Cdk5) may cause oxidative stress by compromising the cellular anti-oxidant defense system. Using novel Cdk5 modulators, we show the mechanism by which Cdk5 can induce oxidative stress in the disease's early stage and cell death in the late stage. Cdk5 dysregulation upon neurotoxic insults results in reactive oxygen species (ROS) accumulation in neuronal cells because of the inactivation of peroxiredoxin I and II. Sole temporal activation of Cdk5 also increases ROS, suggesting its major role in this process. Cdk5 inhibition rescues mitochondrial damage upon neurotoxic insults, thereby revealing Cdk5 as an upstream regulator of mitochondrial dysfunction. As mitochondrial damage results in elevated ROS and Ca(2+) levels, both of which activate Cdk5, we propose that a feedback loop occurs in late stage of AD and leads to cell death (active Cdk5 --> ROS --> excess ROS --> mitochondrial damage --> ROS --> hyperactive Cdk5 --> severe oxidative stress and cell injury --> cell death). Cdk5 inhibition upon neurotoxic insult prevents cell death significantly, supporting this hypothesis. As oxidative stress and mitochondrial dysfunction play pivotal roles in promoting neurodegeneration, Cdk5 could be a viable therapeutic target for AD.


Cyclin-Dependent Kinase 5/metabolism , Energy Metabolism/physiology , Gene Expression Regulation, Enzymologic/genetics , Mitochondria/enzymology , Neurons/metabolism , Oxidative Stress/physiology , Alzheimer Disease/enzymology , Alzheimer Disease/genetics , Alzheimer Disease/physiopathology , Animals , Calcium Signaling/drug effects , Calcium Signaling/physiology , Cell Death/drug effects , Cell Death/physiology , Cells, Cultured , Cyclin-Dependent Kinase 5/antagonists & inhibitors , Energy Metabolism/drug effects , Enzyme Activation/drug effects , Enzyme Activation/physiology , Enzyme Inhibitors/pharmacology , Feedback, Physiological/drug effects , Feedback, Physiological/physiology , Gene Expression Regulation, Enzymologic/drug effects , Genes, cdc/drug effects , Genes, cdc/physiology , Mice , Mitochondria/drug effects , Nerve Degeneration/enzymology , Nerve Degeneration/genetics , Nerve Degeneration/physiopathology , Neurotoxins/pharmacology , Oxidative Stress/drug effects , PC12 Cells , Peroxiredoxins/metabolism , Rats , Reactive Oxygen Species/metabolism , Up-Regulation/drug effects , Up-Regulation/physiology
...