Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
FASEB J ; 37(11): e23218, 2023 11.
Article in English | MEDLINE | ID: mdl-37779443

ABSTRACT

Psychological stress and traumatic brain injury (TBI) result in long-lasting emotional and behavioral impairments in patients. So far, the interaction of psychological stress with TBI not only in the brain but also in peripheral organs is poorly understood. Herein, the impact of acute stress (AS) occurring immediately before TBI is investigated. For this, a mouse model of restraint stress and TBI was employed, and their influence on behavior and gene expression in brain regions, the hypothalamic-pituitary-adrenal (HPA) axis, and peripheral organs was analyzed. Results demonstrate that, compared to single AS or TBI exposure, mice treated with AS prior to TBI showed sex-specific alterations in body weight, memory function, and locomotion. The induction of immediate early genes (IEGs, e.g., c-Fos) by TBI was modulated by previous AS in several brain regions. Furthermore, IEG upregulation along the HPA axis (e.g., pituitary, adrenal glands) and other peripheral organs (e.g., heart) was modulated by AS-TBI interaction. Proteomics of plasma samples revealed proteins potentially mediating this interaction. Finally, the deletion of Atf3 diminished the TBI-induced induction of IEGs in peripheral organs but left them largely unaltered in the brain. In summary, AS immediately before brain injury affects the brain and, to a strong degree, also responses in peripheral organs.


Subject(s)
Brain Injuries, Traumatic , Hypothalamo-Hypophyseal System , Humans , Male , Female , Mice , Animals , Pituitary-Adrenal System , Brain Injuries, Traumatic/metabolism , Pituitary Gland/metabolism , Stress, Psychological/genetics , Stress, Psychological/metabolism , Gene Expression
2.
JCI Insight ; 8(15)2023 08 08.
Article in English | MEDLINE | ID: mdl-37339001

ABSTRACT

Changes in neuronal activity modulate the vulnerability of motoneurons (MNs) in neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS). So far, the molecular basis of neuronal activity's impact in ALS is poorly understood. Herein, we investigated the impact of deleting the neuronal activity-stimulated transcription factor (TF) serum response factor (SRF) in MNs of SOD1G93A mice. SRF was present in vulnerable MMP9+ MNs. Ablation of SRF in MNs induced an earlier disease onset starting around 7-8 weeks after birth, as revealed by enhanced weight loss and decreased motor ability. This earlier disease onset in SRF-depleted MNs was accompanied by a mild elevation of neuroinflammation and neuromuscular synapse degeneration, whereas overall MN numbers and mortality were unaffected. In SRF-deficient mice, MNs showed impaired induction of autophagy-encoding genes, suggesting a potentially new SRF function in transcriptional regulation of autophagy. Complementary, constitutively active SRF-VP16 enhanced autophagy-encoding gene transcription and autophagy progression in cells. Furthermore, SRF-VP16 decreased ALS-associated aggregate induction. Chemogenetic modulation of neuronal activity uncovered SRF as having important TF-mediating activity-dependent effects, which might be beneficial to reduce ALS disease burden. Thus, our data identify SRF as a gene regulator connecting neuronal activity with the cellular autophagy program initiated in degenerating MNs.


Subject(s)
Amyotrophic Lateral Sclerosis , Animals , Mice , Amyotrophic Lateral Sclerosis/genetics , Etoposide , Gene Expression Regulation , Motor Neurons/physiology , Serum Response Factor/genetics
3.
J Neurosci ; 42(12): 2474-2491, 2022 03 23.
Article in English | MEDLINE | ID: mdl-35149515

ABSTRACT

In postmitotic neurons, several tumor suppressor genes (TSGs), including p53, Rb, and PTEN, modulate the axon regeneration success after injury. Particularly, PTEN inhibition is a key driver of successful CNS axon regeneration after optic nerve or spinal cord injury. In contrast, in peripheral neurons, TSG influence in neuronal morphology, physiology, and pathology has not been investigated to the same depth. In this study, we conditionally deleted PTEN from mouse facial motoneurons (Chat-Cre/PtenloxP/loxP ) and analyzed neuronal responses in vivo with or without peripheral facial nerve injury in male and female mice. In uninjured motoneurons, PTEN loss induced somatic, axonal, and nerve hypertrophy, synaptic terminal enlargement and reduction in physiological whisker movement. Despite these morphologic and physiological changes, PTEN deletion positively regulated facial nerve regeneration and recovery of whisker movement after nerve injury. Regenerating PTEN-deficient motoneurons upregulated P-CREB and a signaling pathway involving P-Akt, P-PRAS40, P-mTOR, and P-4EBP1. In aged mice (12 months), PTEN deletion induced hair loss and facial hyperplasia of the epidermis. This suggests a time window in younger mice with PTEN loss stimulating axon growth after injury, however, at the risk of hyperplasia formation at later time points in the old animal. Overall, our data highlight a dual TSG function with PTEN loss impairing physiological neuron function but furthermore underscoring the positive effects of PTEN ablation in axon regeneration also for the PNS.SIGNIFICANCE STATEMENT Tumor suppressor genes (TSGs) restrict cell proliferation and growth. TSG inhibition, including p53 and PTEN, stimulates axon regeneration after CNS injury. In contrast, in PNS axon regeneration, TSGs have not been analyzed in great depth. Herein we show enhanced peripheral axon regeneration after PTEN deletion from facial motoneurons. This invokes a signaling cascade with novel PTEN partners, including CREB and PRAS40. In adult mice, PTEN loss induces hyperplasia of the skin epidermis, suggesting detrimental consequences when reaching adulthood in contrast to a beneficial TSG role for regeneration in young adult mice. Thus, our data highlight the double-edged sword nature of interfering with TSG function.


Subject(s)
Facial Nerve Injuries , Nerve Regeneration , PTEN Phosphohydrolase/metabolism , Animals , Axons/physiology , Facial Nerve Injuries/genetics , Facial Nerve Injuries/pathology , Female , Hyperplasia/pathology , Hypertrophy/pathology , Male , Mice , Motor Neurons/metabolism , Nerve Regeneration/genetics , Tumor Suppressor Protein p53
4.
Cell Rep ; 30(10): 3506-3519.e6, 2020 03 10.
Article in English | MEDLINE | ID: mdl-32160553

ABSTRACT

Genetic variations in TMEM106B, coding for a lysosomal membrane protein, affect frontotemporal lobar degeneration (FTLD) in GRN- (coding for progranulin) and C9orf72-expansion carriers and might play a role in aging. To determine the physiological function of TMEM106B, we generated TMEM106B-deficient mice. These mice develop proximal axonal swellings caused by drastically enlarged LAMP1-positive vacuoles, increased retrograde axonal transport of lysosomes, and accumulation of lipofuscin and autophagosomes. Giant vacuoles specifically accumulate at the distal end and within the axon initial segment, but not in peripheral nerves or at axon terminals, resulting in an impaired facial-nerve-dependent motor performance. These data implicate TMEM106B in mediating the axonal transport of LAMP1-positive organelles in motoneurons and axonal sorting at the initial segment. Our data provide mechanistic insight into how TMEM106B affects lysosomal proteolysis and degradative capacity in neurons.


Subject(s)
Axon Initial Segment/metabolism , Frontotemporal Lobar Degeneration/genetics , Genetic Predisposition to Disease , Lysosomes/metabolism , Membrane Proteins/genetics , Motor Neurons/metabolism , Nerve Tissue Proteins/genetics , Animals , Autophagosomes/metabolism , Autophagosomes/ultrastructure , Axon Initial Segment/ultrastructure , Axonal Transport , Brain Stem/pathology , Cell Nucleus/metabolism , Facial Nerve/pathology , Lysosomes/ultrastructure , Membrane Proteins/deficiency , Mice, Inbred C57BL , Mice, Knockout , Motor Neurons/ultrastructure , Muscles/innervation , Nerve Tissue Proteins/deficiency , Risk Factors
5.
Brain Behav Immun ; 81: 228-246, 2019 10.
Article in English | MEDLINE | ID: mdl-31207335

ABSTRACT

Traumatic brain injury (TBI) and ethanol intoxication (EI) frequently coincide, particularly in young subjects. However, the mechanisms of their interaction remain poorly understood. Among other pathogenic pathways, TBI induces glial activation and neuroinflammation in the hippocampus, resulting in acute and chronic hippocampal dysfunction. In this regard, we investigated the role of EI affecting these responses unfolding after TBI. We used a blunt, weight-drop approach to model TBI in mice. Male mice were pre-administered with ethanol or vehicle to simulate EI. The neuroinflammatory response in the hippocampus was assessed by monitoring the expression levels of >20 cytokines, the phosphorylation status of transcription factors and the phenotype of microglia and astrocytes. We used AS1517499, a brain-permeable STAT6 inhibitor, to elucidate the role of this pathway in the EI/TBI interaction. We showed that TBI causes the elevation of IL-33, IL-1ß, IL-38, TNF-α, IFN-α, IL-19 in the hippocampus at 3 h time point and concomitant EI results in the dose-dependent downregulation of IL-33, IL-1ß, IL-38, TNF-α and IL-19 (but not of IFN-α) and in the selective upregulation of IL-13 and IL-12. EI is associated with the phosphorylation of STAT6 and the transcription of STAT6-controlled genes. Moreover, ethanol-induced STAT6 phosphorylation and transcriptional activation can be recapitulated in vitro by concomitant exposure of neurons to ethanol, depolarization and inflammatory stimuli (simulating the acute trauma). Acute STAT6 inhibition prevents the effects of EI on IL-33 and TNF-α, but not on IL-13 and negates acute EI beneficial effects on TBI-associated neurological impairment. Additionally, EI is associated with reduced microglial activation and astrogliosis as well as preserved synaptic density and baseline neuronal activity 7 days after TBI and all these effects are prevented by acute administration of the STAT6 inhibitor concomitant to EI. EI concomitant to TBI exerts significant immunomodulatory effects on cytokine induction and microglial activation, largely through the activation of STAT6 pathway, ultimately with beneficial outcomes.


Subject(s)
Brain Injuries, Traumatic/metabolism , Ethanol/pharmacology , STAT6 Transcription Factor/metabolism , Animals , Brain/metabolism , Brain/pathology , Brain Injuries, Traumatic/immunology , Brain Injuries, Traumatic/pathology , Cytokines/metabolism , Disease Models, Animal , Macrophage Activation/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Microglia/metabolism , Microglia/pathology , Neuroimmunomodulation/drug effects , Neurons/metabolism , Neurons/pathology , STAT6 Transcription Factor/immunology , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/metabolism
6.
Proc Natl Acad Sci U S A ; 116(3): 880-889, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30598445

ABSTRACT

Serum response factor (SRF) mediates immediate early gene (IEG) and cytoskeletal gene expression programs in almost any cell type. So far, SRF transcriptional dynamics have not been investigated at single-molecule resolution. We provide a study of single Halo-tagged SRF molecules in fibroblasts and primary neurons. In both cell types, individual binding events of SRF molecules segregated into three chromatin residence time regimes, short, intermediate, and long binding, indicating a cell type-independent SRF property. The chromatin residence time of the long bound fraction was up to 1 min in quiescent cells and significantly increased upon stimulation. Stimulation also enhanced the long bound SRF fraction at specific timepoints (20 and 60 min) in both cell types. These peaks correlated with activation of the SRF cofactors MRTF-A and MRTF-B (myocardin-related transcription factors). Interference with signaling pathways and cofactors demonstrated modulation of SRF chromatin occupancy by actin signaling, MAP kinases, and MRTFs.


Subject(s)
Chromatin/metabolism , Serum Response Factor/metabolism , Actins/metabolism , Animals , Fibroblasts/metabolism , MAP Kinase Signaling System , Mice , NIH 3T3 Cells , Neurons/metabolism , Single Molecule Imaging , Trans-Activators/metabolism , Transcription Factors/metabolism
7.
J Neurotrauma ; 35(19): 2317-2329, 2018 10 01.
Article in English | MEDLINE | ID: mdl-29463176

ABSTRACT

Traumatic brain injury (TBI) induces a neuroinflammatory response resulting in astrocyte and microglia activation at the lesion site. This involves upregulation of neuroinflammatory genes, including chemokines and interleukins. However, so far, there is lack of knowledge on transcription factors (TFs) modulating this TBI-associated gene expression response. Herein, we analyzed activating transcription factor 3 (ATF3), a TF encoding a regeneration-associated gene (RAG) predominantly studied in peripheral nervous system (PNS) injury. ATF3 contributes to PNS axon regeneration and was shown before to regulate inflammatory processes in other injury models. In contrast to PNS injury, data on ATF3 in central nervous system (CNS) injury are sparse. We used Atf3 mouse mutants and a closed-head weight-drop-based TBI model in adult mice to target the rostrolateral cortex resulting in moderate injury severity. Post-TBI, ATF3 was upregulated already at early time points (i.e,. 1-4 h) post-injury in the brain. Mortality and weight loss upon TBI were slightly elevated in Atf3 mutants. ATF3 deficiency enhanced TBI-induced paresis and hematoma formation, suggesting that ATF3 limits these injury outcomes in wild-type mice. Next, we analyzed TBI-associated RAG and inflammatory gene expression in the cortical impact area. In contrast to the PNS, only some RAGs (Atf3, Timp1, and Sprr1a) were induced by TBI, and, surprisingly, some RAG encoding neuropeptides were downregulated. Notably, we identified ATF3 as TF-regulating proneuroinflammatory gene expression, including CCL and CXCL chemokines (Ccl2, Ccl3, Ccl4, and Cxcl1) and lipocalin. In Atf3 mutant mice, mRNA abundance was further enhanced upon TBI compared to wild-type mice, suggesting immune gene repression by wild-type ATF3. In accord, more immune cells were present in the lesion area of ATF3-deficient mice. Overall, we identified ATF3 as a new TF-mediating TBI-associated CNS inflammatory responses.


Subject(s)
Activating Transcription Factor 3/immunology , Brain Injuries, Traumatic/immunology , Brain Injuries, Traumatic/pathology , Inflammation/immunology , Inflammation/pathology , Animals , Female , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains
8.
Exp Neurol ; 302: 34-45, 2018 04.
Article in English | MEDLINE | ID: mdl-29306704

ABSTRACT

Ethanol intoxication is a risk factor for traumatic brain injury (TBI) but clinical evidence suggests that it may actually improve the prognosis of intoxicated TBI patients. We have employed a closed, weight-drop TBI model of different severity (2cm or 3cm falling height), preceded (-30min) or followed (+20min) by ethanol administration (5g/Kg). This protocol allows us to study the interaction of binge ethanol intoxication in TBI, monitoring behavioral changes, histological responses and the transcriptional regulation of a series of activity-regulated genes (immediate early genes, IEGs). We demonstrate that ethanol pretreatment before moderate TBI (2cm) significantly reduces neurological impairment and accelerates recovery. In addition, better preservation of neuronal numbers and cFos+cells was observed 7days after TBI. At transcriptional level, ethanol reduced the upregulation of a subset of IEGs encoding for transcription factors such as Atf3, c-Fos, FosB, Egr1, Egr3 and Npas4 but did not affect the upregulation of others (e.g. Gadd45b and Gadd45c). While a subset of IEGs encoding for effector proteins (such as Bdnf, InhbA and Dusp5) were downregulated by ethanol, others (such as Il-6) were unaffected. Notably, the majority of genes were sensitive to ethanol only when administered before TBI and not afterwards (the exceptions being c-Fos, Egr1 and Dusp5). Furthermore, while severe TBI (3cm) induced a qualitatively similar (but quantitatively larger) transcriptional response to moderate TBI, it was no longer sensitive to ethanol pretreatment. Thus, we have shown that a subset of the TBI-induced transcriptional responses were sensitive to ethanol intoxication at the instance of trauma (ultimately resulting in beneficial outcomes) and that the effect of ethanol was restricted to a certain time window (pre TBI treatment) and to TBI severity (moderate). This information could be critical for the translational value of ethanol in TBI and for the design of clinical studies aimed at disentangling the role of ethanol intoxication in TBI.


Subject(s)
Alcoholic Intoxication/prevention & control , Brain Injuries, Traumatic/chemically induced , Brain Injuries, Traumatic/prevention & control , Central Nervous System Depressants/administration & dosage , Ethanol/administration & dosage , Transcription Factors/metabolism , Analysis of Variance , Animals , Central Nervous System Depressants/blood , Dose-Response Relationship, Drug , Ethanol/blood , Exploratory Behavior/drug effects , Male , Mice , Neurologic Examination , RNA, Messenger/metabolism , Transcription Factors/genetics , Transcription, Genetic/drug effects
9.
Mol Brain ; 10(1): 30, 2017 07 17.
Article in English | MEDLINE | ID: mdl-28716058

ABSTRACT

A hallmark of temporal lobe epilepsy (TLE) is hippocampal neuronal demise and aberrant mossy fiber sprouting. In addition, unrestrained neuronal activity in TLE patients induces gene expression including immediate early genes (IEGs) such as Fos and Egr1.We employed the mouse pilocarpine model to analyze the transcription factor (TF) serum response factor (SRF) in epileptogenesis, seizure induced histopathology and IEG induction. SRF is a neuronal activity regulated TF stimulating IEG expression as well as nerve fiber growth and guidance. Adult conditional SRF deficient mice (Srf CaMKCreERT2 ) were more refractory to initial status epilepticus (SE) acquisition. Further, SRF deficient mice developed more spontaneous recurrent seizures (SRS). Genome-wide transcriptomic analysis uncovered a requirement of SRF for SE and SRS induced IEG induction (e.g. Fos, Egr1, Arc, Npas4, Btg2, Atf3). SRF was required for epilepsy associated neurodegeneration, mossy fiber sprouting and inflammation. We uncovered MAP kinase signaling as SRF target during epilepsy. Upon SRF ablation, seizure evoked induction of dual specific phosphatases (Dusp5 and Dusp6) was reduced. Lower expression of these negative ERK kinase regulators correlated with altered P-ERK levels in epileptic Srf mutant animals.Overall, this study uncovered an SRF contribution to several processes of epileptogenesis in the pilocarpine model.


Subject(s)
Epilepsy/genetics , Epilepsy/pathology , Hippocampus/pathology , Nerve Net/pathology , Seizures/genetics , Seizures/pathology , Serum Response Factor/metabolism , Transcription, Genetic , Animals , Disease Models, Animal , Dual-Specificity Phosphatases/metabolism , Epilepsy/chemically induced , Gene Expression Regulation , MAP Kinase Signaling System , Mice, Inbred C57BL , Mossy Fibers, Hippocampal/metabolism , Mossy Fibers, Hippocampal/pathology , Nerve Net/metabolism , Neurons/metabolism , Neurons/pathology , Pilocarpine , Reproducibility of Results
10.
Open Biol ; 6(8)2016 08.
Article in English | MEDLINE | ID: mdl-27581653

ABSTRACT

Axon injury in the peripheral nervous system (PNS) induces a regeneration-associated gene (RAG) response. Atf3 (activating transcription factor 3) is such a RAG and ATF3's transcriptional activity might induce 'effector' RAGs (e.g. small proline rich protein 1a (Sprr1a), Galanin (Gal), growth-associated protein 43 (Gap43)) facilitating peripheral axon regeneration. We provide a first analysis of Atf3 mouse mutants in peripheral nerve regeneration. In Atf3 mutant mice, facial nerve regeneration and neurite outgrowth of adult ATF3-deficient primary dorsal root ganglia neurons was decreased. Using genome-wide transcriptomics, we identified a neuropeptide-encoding RAG cluster (vasoactive intestinal peptide (Vip), Ngf, Grp, Gal, Pacap) regulated by ATF3. Exogenous administration of neuropeptides enhanced neurite growth of Atf3 mutant mice suggesting that these molecules might be effector RAGs of ATF3's pro-regenerative function. In addition to the induction of growth-promoting molecules, we present data that ATF3 suppresses growth-inhibiting molecules such as chemokine (C-C motif) ligand 2. In summary, we show a pro-regenerative ATF3 function during PNS nerve regeneration involving transcriptional activation of a neuropeptide-encoding RAG cluster. ATF3 is a general injury-inducible factor, therefore ATF3-mediated mechanisms identified herein might apply to other cell and injury types.


Subject(s)
Activating Transcription Factor 3/genetics , Axons/pathology , Mutation , Neuropeptides/genetics , Peripheral Nerve Injuries/pathology , Animals , Disease Models, Animal , Ganglia, Spinal/pathology , Gene Expression Profiling , Gene Regulatory Networks , Mice , Nerve Growth Factor/genetics , Nerve Regeneration , Peripheral Nerve Injuries/genetics , Pituitary Adenylate Cyclase-Activating Polypeptide/genetics
11.
Glia ; 63(6): 958-76, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25639799

ABSTRACT

In multiple sclerosis (MS), neurons in addition to inflammatory cells are now considered to mediate disease origin and progression. So far, molecular and cellular mechanisms of neuronal MS contributions are poorly understood. Herein we analyzed whether neuron-restricted signaling by the neuroprotective transcription factor serum response factor (SRF) modulates de- and remyelination in a rodent MS model. In the mouse cuprizone model, neuron- (Srf (flox/flox;CaMKCreERT2)) but not glia-specific (Srf (flox/flox;PlpCreERT2)) SRF depletion impaired demyelination suggesting impaired debris clearance by astrocytes and microglia. This supports an important role of SRF expression in neurons but not oligodendrocytes in de- and remyelination. During remyelination, NG2- and OLIG2-positive cells of the oligodendrocyte lineage as well as de novo mRNA synthesis of myelin genes were also reduced in neuron-specific Srf mutants. Using the stripe assay, we demonstrate that cortices of cuprizone-fed wild-type mice elicited astrocyte and microglia activation whereas this was abrogated in cuprizone-fed neuron-specific Srf mutants. We identified CCL chemokines (e.g. CCL2) as neuron-derived SRF-regulated paracrine signals rescuing immune cell activation upon neuronal SRF deletion. In summary, we uncovered important roles of neurons and neuronally expressed SRF in MS associated de- and remyelination.


Subject(s)
Multiple Sclerosis/physiopathology , Myelin Sheath/physiology , Neurons/metabolism , Serum Response Factor/metabolism , Animals , Astrocytes/pathology , Astrocytes/physiology , Cells, Cultured , Chemokine CCL2/metabolism , Cuprizone , Disease Models, Animal , Male , Mice, Inbred C57BL , Mice, Transgenic , Microglia/pathology , Microglia/physiology , Multiple Sclerosis/pathology , Myelin Sheath/metabolism , Myelin Sheath/pathology , Neurons/pathology , Oligodendroglia/pathology , Oligodendroglia/physiology , RNA, Messenger/metabolism , Serum Response Factor/genetics
12.
Histochem Cell Biol ; 141(2): 123-35, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24091797

ABSTRACT

Compared to the cytoplasmic F-actin abundance in cells, nuclear F-actin levels are generally quite low. However, nuclear actin is present in certain cell types including oocytes and under certain cellular conditions including stress or serum stimulation. Currently, the architecture and polymerization status of nuclear actin networks has not been analyzed in great detail. In this study, we investigated the architecture and functions of such nuclear actin networks. We generated nuclear actin polymers by overexpression of actin proteins fused to a nuclear localization signal (NLS). Raising nuclear abundance of a NLS wild-type actin, we observed phalloidin- and LifeAct-positive actin bundles forming a nuclear cytoskeletal network consisting of curved F-actin. In contrast, a polymer-stabilizing actin mutant (NLS-G15S-actin) deficient in interacting with the actin-binding protein cofilin generated a nuclear actin network reminiscent of straight stress fiber-like microfilaments in the cytoplasm. We provide a first electron microscopic description of such nuclear actin polymers suggesting bundling of actin filaments. Employing different cell types from various species including neurons, we show that the morphology of and potential to generate nuclear actin are conserved. Finally, we demonstrate that nuclear actin affects cell function including morphology, serum response factor-mediated gene expression, and herpes simplex virus infection. Our data suggest that actin is able to form filamentous structures inside the nucleus, which share architectural and functional similarities with the cytoplasmic F-actin.


Subject(s)
Actins/genetics , Cell Nucleus/metabolism , Gene Expression , Mutant Proteins/genetics , Actins/metabolism , Actins/ultrastructure , Cell Line , HEK293 Cells , Humans , Immunohistochemistry , Mutant Proteins/metabolism
13.
J Neurosci ; 33(48): 18836-48, 2013 Nov 27.
Article in English | MEDLINE | ID: mdl-24285890

ABSTRACT

Axonal injury generates growth inert retraction bulbs with dynamic cytoskeletal properties that are severely compromised. Conversion of "frozen" retraction bulbs into actively progressing growth cones is a major aim in axon regeneration. Here we report that murine serum response factor (SRF), a gene regulator linked to the actin cytoskeleton, modulates growth cone actin dynamics during axon regeneration. In regeneration-competent facial motoneurons, Srf deletion inhibited axonal regeneration. In wild-type mice after nerve injury, SRF translocated from the nucleus to the cytoplasm, suggesting a cytoplasmic SRF function in axonal regeneration. Indeed, adenoviral overexpression of cytoplasmic SRF (SRF-ΔNLS-GFP) stimulated axonal sprouting and facial nerve regeneration in vivo. In primary central and peripheral neurons, SRF-ΔNLS-GFP stimulated neurite outgrowth, branch formation, and growth cone morphology. Furthermore, we uncovered a link between SRF and the actin-severing factor cofilin during axonal regeneration in vivo. Facial nerve axotomy increased the total cofilin abundance and also nuclear localization of phosphorylated cofilin in a subpopulation of lesioned motoneurons. This cytoplasmic-to-nucleus translocation of P-cofilin upon axotomy was reduced in motoneurons expressing SRF-ΔNLS-GFP. Finally, we demonstrate that cytoplasmic SRF and cofilin formed a reciprocal regulatory unit. Overexpression of cytoplasmic SRF reduced cofilin phosphorylation and vice versa: overexpression of cofilin inhibited SRF phosphorylation. Therefore, a regulatory loop consisting of SRF and cofilin might take part in reactivating actin dynamics in growth-inert retraction bulbs and facilitating axon regeneration.


Subject(s)
Actin Depolymerizing Factors/physiology , Axons/drug effects , Cytoplasm/metabolism , Nerve Regeneration/drug effects , Serum Response Factor/pharmacology , Actins/metabolism , Animals , Axotomy , Cytoplasm/drug effects , Facial Nerve/physiology , Female , Green Fluorescent Proteins , Male , Mice , Peripheral Nerves/cytology , Peripheral Nerves/drug effects , Phosphorylation , Polymerase Chain Reaction , Subcellular Fractions/metabolism
14.
J Neuroinflammation ; 9: 78, 2012 Apr 26.
Article in English | MEDLINE | ID: mdl-22537405

ABSTRACT

BACKGROUND: The transcription factor SRF (serum response factor) mediates neuronal survival in vitro. However, data available so far suggest that SRF is largely dispensable for neuron survival during physiological brain function. FINDINGS: Here, we demonstrate that upon neuronal injury, that is facial nerve transection, constitutively-active SRF-VP16 enhances motorneuron survival. SRF-VP16 suppressed active caspase 3 abundance in vitro and enhanced neuron survival upon camptothecin induced apoptosis. Following nerve fiber injury in vitro, SRF-VP16 improved survival of neurons and re-growth of severed neurites. Further, SRF-VP16 enhanced immune responses (that is microglia and T cell activation) associated with neuronal injury in vivo. Genome-wide transcriptomics identified target genes associated with axonal injury and modulated by SRF-VP16. CONCLUSION: In sum, this is a first report describing a neuronal injury-related survival function for SRF.


Subject(s)
Axons/pathology , Facial Nerve Injuries/pathology , Neurons/pathology , Peripheral Nerve Injuries/pathology , Serum Response Factor/physiology , Animals , Axons/physiology , Cell Survival/genetics , Disease Models, Animal , Facial Nerve Injuries/genetics , Mice , Mice, Knockout , Neurons/physiology , Peripheral Nerve Injuries/genetics , Serum Response Factor/deficiency , Serum Response Factor/genetics
15.
Nat Neurosci ; 12(4): 418-27, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19270689

ABSTRACT

In neurons, serum response factor (SRF)-directed transcription regulates migration, axon pathfinding and synapse function. We found that forebrain-specific, neuron-restricted SRF ablation in mice elevated oligodendrocyte precursors while inhibiting terminal oligodendrocyte differentiation. Myelin gene and protein expression were downregulated and we observed a lack of oligodendrocytes in mixed neuron/glia and oligodendrocyte-enriched cultures derived from Srf(-/-) mutants. Ultrastructural inspection revealed myelination defects and axonal degeneration in Srf(-/-) mutants. Consistent with our finding that neuronal SRF depletion impaired oligodendrocyte fate in a non-cell autonomous manner, neuron-restricted expression of constitutively active SRF-VP16 affected neighboring oligodendrocyte maturation. Genome-wide transcriptomics identified candidate genes for paracrine regulation of oligodendrocyte development, including connective tissue growth factor (CTGF), whose expression is repressed by SRF. Adenovirus-mediated CTGF expression in vivo revealed that CTGF blocks excessive oligodendrocyte differentiation. In vitro, CTGF-mediated inhibition of oligodendrocyte maturation involved sequestration and thereby counteraction of insulin growth factor 1-stimulated oligodendrocyte differentiation.


Subject(s)
Gene Expression/physiology , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Paracrine Communication/physiology , Serum Response Factor/metabolism , Animals , Animals, Newborn , Axons/metabolism , Axons/ultrastructure , Brain/cytology , Brain/ultrastructure , Cell Differentiation/physiology , Cells, Cultured , Coculture Techniques/methods , Connective Tissue Growth Factor/genetics , Embryo, Mammalian , Gene Expression/genetics , Gene Expression Regulation, Developmental/genetics , Green Fluorescent Proteins/genetics , Humans , Insulin-Like Growth Factor I/metabolism , Mice , Mice, Knockout , Myelin Sheath/metabolism , Myelin Sheath/ultrastructure , Nerve Tissue Proteins/genetics , Neuroglia/physiology , Neurons/ultrastructure , Oligodendroglia/metabolism , Paracrine Communication/genetics , Serum Response Factor/deficiency
SELECTION OF CITATIONS
SEARCH DETAIL
...