Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 112
Filter
Add more filters

Country/Region as subject
Publication year range
1.
EMBO J ; 38(13): e101032, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31268609

ABSTRACT

The molecular mechanisms discriminating between regenerative failure and success remain elusive. While a regeneration-competent peripheral nerve injury mounts a regenerative gene expression response in bipolar dorsal root ganglia (DRG) sensory neurons, a regeneration-incompetent central spinal cord injury does not. This dichotomic response offers a unique opportunity to investigate the fundamental biological mechanisms underpinning regenerative ability. Following a pharmacological screen with small-molecule inhibitors targeting key epigenetic enzymes in DRG neurons, we identified HDAC3 signalling as a novel candidate brake to axonal regenerative growth. In vivo, we determined that only a regenerative peripheral but not a central spinal injury induces an increase in calcium, which activates protein phosphatase 4 that in turn dephosphorylates HDAC3, thus impairing its activity and enhancing histone acetylation. Bioinformatics analysis of ex vivo H3K9ac ChIPseq and RNAseq from DRG followed by promoter acetylation and protein expression studies implicated HDAC3 in the regulation of multiple regenerative pathways. Finally, genetic or pharmacological HDAC3 inhibition overcame regenerative failure of sensory axons following spinal cord injury. Together, these data indicate that PP4-dependent HDAC3 dephosphorylation discriminates between axonal regeneration and regenerative failure.


Subject(s)
Ganglia, Spinal/physiology , Histone Deacetylases/metabolism , Peripheral Nerve Injuries/metabolism , Phosphoprotein Phosphatases/metabolism , Small Molecule Libraries/pharmacology , Animals , Axons , Cells, Cultured , Disease Models, Animal , Epigenesis, Genetic/drug effects , Female , Male , Mice , Nerve Regeneration , Phosphorylation/drug effects , Signal Transduction
2.
Proc Natl Acad Sci U S A ; 117(52): 33597-33607, 2020 12 29.
Article in English | MEDLINE | ID: mdl-33318207

ABSTRACT

Axon injury is a hallmark of many neurodegenerative diseases, often resulting in neuronal cell death and functional impairment. Dual leucine zipper kinase (DLK) has emerged as a key mediator of this process. However, while DLK inhibition is robustly protective in a wide range of neurodegenerative disease models, it also inhibits axonal regeneration. Indeed, there are no genetic perturbations that are known to both improve long-term survival and promote regeneration. To identify such a neuroprotective target, we conducted a set of complementary high-throughput screens using a protein kinase inhibitor library in human stem cell-derived retinal ganglion cells (hRGCs). Overlapping compounds that promoted both neuroprotection and neurite outgrowth were bioinformatically deconvoluted to identify specific kinases that regulated neuronal death and axon regeneration. This work identified the role of germinal cell kinase four (GCK-IV) kinases in cell death and additionally revealed their unexpected activity in suppressing axon regeneration. Using an adeno-associated virus (AAV) approach, coupled with genome editing, we validated that GCK-IV kinase knockout improves neuronal survival, comparable to that of DLK knockout, while simultaneously promoting axon regeneration. Finally, we also found that GCK-IV kinase inhibition also prevented the attrition of RGCs in developing retinal organoid cultures without compromising axon outgrowth, addressing a major issue in the field of stem cell-derived retinas. Together, these results demonstrate a role for the GCK-IV kinases in dissociating the cell death and axonal outgrowth in neurons and their druggability provides for therapeutic options for neurodegenerative diseases.


Subject(s)
Axons/enzymology , Axons/pathology , Central Nervous System/pathology , Germinal Center Kinases/metabolism , Nerve Regeneration , Animals , Base Sequence , CRISPR-Cas Systems/genetics , Cell Death/drug effects , Cell Survival/drug effects , Dependovirus/metabolism , Disease Models, Animal , Humans , Mice, Inbred C57BL , Nerve Regeneration/drug effects , Neuronal Outgrowth/drug effects , Optic Nerve Injuries/metabolism , Optic Nerve Injuries/pathology , Organoids/metabolism , Protein Kinase Inhibitors/pharmacology , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/metabolism , Signal Transduction/drug effects
3.
J Neurosci ; 41(1): 3-10, 2021 01 06.
Article in English | MEDLINE | ID: mdl-33408132

ABSTRACT

In 1981, I published a paper in the first issue of the Journal of Neuroscience with my postdoctoral mentor, Alan Pearlman. It reported a quantitative analysis of the receptive field properties of neurons in reeler mouse visual cortex and the surprising conclusion that although the neuronal somas were strikingly malpositioned, their receptive fields were unchanged. This suggested that in mouse cortex at least, neuronal circuits have very robust systems in place to ensure the proper formation of connections. This had the unintended consequence of transforming me from an electrophysiologist into a cellular and molecular neuroscientist who studied cell adhesion molecules and the molecular mechanisms they use to regulate axon growth. It took me a surprisingly long time to appreciate that your science is driven by the people around you and by the technologies that are locally available. As a professional puzzler, I like all different kinds of puzzles, but the most fun puzzles involve playing with other puzzlers. This is my story of learning how to find like-minded puzzlers to solve riddles about axon growth and regeneration.


Subject(s)
Axons , Neurology/history , Visual Cortex/growth & development , Visual Cortex/physiology , Animals , History, 20th Century , Mice , Mice, Neurologic Mutants , Neurons/physiology , Visual Pathways/physiology
5.
Mol Cell Neurosci ; 92: 114-127, 2018 10.
Article in English | MEDLINE | ID: mdl-30077771

ABSTRACT

Axon regeneration is a necessary step toward functional recovery after spinal cord injury. The AP-1 transcription factor c-Jun has long been known to play an important role in directing the transcriptional response of Dorsal Root Ganglion (DRG) neurons to peripheral axotomy that results in successful axon regeneration. Here we performed ChIPseq for Jun in mouse DRG neurons after a sciatic nerve crush or sham surgery in order to measure the changes in Jun's DNA binding in response to peripheral axotomy. We found that the majority of Jun's injury-responsive changes in DNA binding occur at putative enhancer elements, rather than proximal to transcription start sites. We also used a series of single polypeptide chain tandem transcription factors to test the effects of different Jun-containing dimers on neurite outgrowth in DRG, cortical and hippocampal neurons. These experiments demonstrated that dimers composed of Jun and Atf3 promoted neurite outgrowth in rat CNS neurons as well as mouse DRG neurons. Our work provides new insight into the mechanisms underlying Jun's role in axon regeneration.


Subject(s)
Neuronal Outgrowth , Protein Multimerization , Proto-Oncogene Proteins c-jun/metabolism , Activating Transcription Factor 3/metabolism , Animals , Brain/cytology , Brain/metabolism , Cells, Cultured , Enhancer Elements, Genetic , Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Neurons/cytology , Neurons/metabolism , Protein Binding , Rats , Rats, Sprague-Dawley
6.
J Neurosci ; 37(30): 7079-7095, 2017 07 26.
Article in English | MEDLINE | ID: mdl-28626016

ABSTRACT

The mammalian target of rapamycin (mTOR) positively regulates axon growth in the mammalian central nervous system (CNS). Although axon regeneration and functional recovery from CNS injuries are typically limited, knockdown or deletion of PTEN, a negative regulator of mTOR, increases mTOR activity and induces robust axon growth and regeneration. It has been suggested that inhibition of S6 kinase 1 (S6K1, gene symbol: RPS6KB1), a prominent mTOR target, would blunt mTOR's positive effect on axon growth. In contrast to this expectation, we demonstrate that inhibition of S6K1 in CNS neurons promotes neurite outgrowth in vitro by twofold to threefold. Biochemical analysis revealed that an mTOR-dependent induction of PI3K signaling is involved in mediating this effect of S6K1 inhibition. Importantly, treating female mice in vivo with PF-4708671, a selective S6K1 inhibitor, stimulated corticospinal tract regeneration across a dorsal spinal hemisection between the cervical 5 and 6 cord segments (C5/C6), increasing axon counts for at least 3 mm beyond the injury site at 8 weeks after injury. Concomitantly, treatment with PF-4708671 produced significant locomotor recovery. Pharmacological targeting of S6K1 may therefore constitute an attractive strategy for promoting axon regeneration following CNS injury, especially given that S6K1 inhibitors are being assessed in clinical trials for nononcological indications.SIGNIFICANCE STATEMENT Despite mTOR's well-established function in promoting axon regeneration, the role of its downstream target, S6 kinase 1 (S6K1), has been unclear. We used cellular assays with primary neurons to demonstrate that S6K1 is a negative regulator of neurite outgrowth, and a spinal cord injury model to show that it is a viable pharmacological target for inducing axon regeneration. We provide mechanistic evidence that S6K1's negative feedback to PI3K signaling is involved in axon growth inhibition, and show that phosphorylation of S6K1 is a more appropriate regeneration indicator than is S6 phosphorylation.


Subject(s)
Axons/metabolism , Imidazoles/administration & dosage , Piperazines/administration & dosage , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/enzymology , Spinal Cord Regeneration/drug effects , TOR Serine-Threonine Kinases/metabolism , Animals , Cells, Cultured , Drug Delivery Systems , Gene Expression Regulation, Enzymologic/physiology , Male , Mice , Mice, Inbred C57BL , Molecular Targeted Therapy , Neuronal Outgrowth/drug effects , Protein Binding , Ribosomal Protein S6 Kinases, 90-kDa/antagonists & inhibitors , Substrate Specificity , Treatment Outcome
7.
Mol Cell Neurosci ; 80: 161-169, 2017 04.
Article in English | MEDLINE | ID: mdl-27444126

ABSTRACT

High-throughput, target-based screening techniques have been utilized extensively for drug discovery in the past several decades. However, the need for more predictive in vitro models of in vivo disease states has generated a shift in strategy towards phenotype-based screens. Phenotype based screens are particularly valuable in studying complex conditions such as CNS injury and degenerative disease, as many factors can contribute to a specific cellular response. In this review, we will discuss different screening frameworks and their relative utility in examining mechanisms of neurodegeneration and axon regrowth, particularly in cell-based in vitro disease models.


Subject(s)
Drug Discovery , Drug Evaluation, Preclinical , Nerve Degeneration/drug therapy , Nerve Regeneration/drug effects , Animals , Humans , Phenotype
9.
Mol Cell Neurosci ; 59: 97-105, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24521823

ABSTRACT

A number of genes regulate regeneration of peripheral axons, but their ability to drive axon growth and regeneration in the central nervous system (CNS) remains largely untested. To address this question we overexpressed eight transcription factors and one small GTPase alone and in pairwise combinations to test whether combinatorial overexpression would have a synergistic impact on CNS neuron neurite growth. The Jun oncogene/signal transducer and activator of transcription 6 (JUN/STAT6) combination increased neurite growth in dissociated cortical neurons and in injured cortical slices. In injured cortical slices, JUN overexpression increased axon growth to a similar extent as JUN and STAT6 together. Interestingly, JUN overexpression was not associated with increased growth associated protein 43 (GAP43) or integrin alpha 7 (ITGA7) expression, though these are predicted transcriptional targets. This study demonstrates that JUN overexpression in cortical neurons stimulates axon growth, but does so independently of changes in expression of genes thought to be critical for JUNs effects on axon growth. We conclude that JUN activity underlies this CNS axonal growth response, and that it is mechanistically distinct from peripheral regeneration responses, in which increases in JUN expression coincide with increases in GAP43 expression.


Subject(s)
Axons/metabolism , Cerebral Cortex/growth & development , Oncogene Protein p65(gag-jun)/metabolism , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Axons/physiology , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , GAP-43 Protein/genetics , GAP-43 Protein/metabolism , Gene Expression Regulation, Developmental , HEK293 Cells , Humans , Integrin alpha Chains/genetics , Integrin alpha Chains/metabolism , Nerve Regeneration , Neurogenesis , Oncogene Protein p65(gag-jun)/genetics , Rats , Rats, Sprague-Dawley , STAT6 Transcription Factor/genetics , STAT6 Transcription Factor/metabolism
10.
Proc Natl Acad Sci U S A ; 109(19): 7517-22, 2012 May 08.
Article in English | MEDLINE | ID: mdl-22529377

ABSTRACT

Axon regeneration in the central nervous system normally fails, in part because of a developmental decline in the intrinsic ability of CNS projection neurons to extend axons. Members of the KLF family of transcription factors regulate regenerative potential in developing CNS neurons. Expression of one family member, KLF7, is down-regulated developmentally, and overexpression of KLF7 in cortical neurons in vitro promotes axonal growth. To circumvent difficulties in achieving high neuronal expression of exogenous KLF7, we created a chimera with the VP16 transactivation domain, which displayed enhanced neuronal expression compared with the native protein while maintaining transcriptional activation and growth promotion in vitro. Overexpression of VP16-KLF7 overcame the developmental loss of regenerative ability in cortical slice cultures. Adult corticospinal tract (CST) neurons failed to up-regulate KLF7 in response to axon injury, and overexpression of VP16-KLF7 in vivo promoted both sprouting and regenerative axon growth in the CST of adult mice. These findings identify a unique means of promoting CST axon regeneration in vivo by reengineering a developmentally down-regulated, growth-promoting transcription factor.


Subject(s)
Axons/physiology , Kruppel-Like Transcription Factors/metabolism , Nerve Regeneration/physiology , Pyramidal Tracts/physiology , Animals , Axons/metabolism , Cells, Cultured , Etoposide , Female , Gene Expression , Genetic Engineering , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Herpes Simplex Virus Protein Vmw65/genetics , Humans , Immunohistochemistry , Kruppel-Like Transcription Factors/genetics , Luminescent Measurements/methods , Mice , Mice, Inbred C57BL , Mutation , Nerve Regeneration/genetics , Neurites/metabolism , Neurites/physiology , Neurons/cytology , Neurons/metabolism , Neurons/physiology , Pyramidal Tracts/cytology , Pyramidal Tracts/metabolism , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Spinal Cord Injuries/genetics , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/physiopathology , Transcriptional Activation
11.
J Infect Dis ; 208(11): 1914-22, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-23901083

ABSTRACT

BACKGROUND: Amphotericin B (AmB), the most effective drug against leishmaniasis, has serious toxicity. As Leishmania species are obligate intracellular parasites of antigen presenting cells (APC), an immunopotentiating APC-specific AmB nanocarrier would be ideally suited to reduce the drug dosage and regimen requirements in leishmaniasis treatment. Here, we report a nanocarrier that results in effective treatment shortening of cutaneous leishmaniasis in a mouse model, while also enhancing L. major specific T-cell immune responses in the infected host. METHODS: We used a Pan-DR-binding epitope (PADRE)-derivatized-dendrimer (PDD), complexed with liposomal amphotericin B (LAmB) in an L. major mouse model and analyzed the therapeutic efficacy of low-dose PDD/LAmB vs full dose LAmB. RESULTS: PDD was shown to escort LAmB to APCs in vivo, enhanced the drug efficacy by 83% and drug APC targeting by 10-fold and significantly reduced parasite burden and toxicity. Fortuitously, the PDD immunopotentiating effect significantly enhanced parasite-specific T-cell responses in immunocompetent infected mice. CONCLUSIONS: PDD reduced the effective dose and toxicity of LAmB and resulted in elicitation of strong parasite specific T-cell responses. A reduced effective therapeutic dose was achieved by selective LAmB delivery to APC, bypassing bystander cells, reducing toxicity and inducing antiparasite immunity.


Subject(s)
Amphotericin B/administration & dosage , Antiprotozoal Agents/administration & dosage , Dendrimers/administration & dosage , Leishmania major/drug effects , Leishmaniasis, Cutaneous/drug therapy , Malaria Vaccines/administration & dosage , Adaptive Immunity , Amphotericin B/toxicity , Animals , Antigen-Presenting Cells/immunology , Antiprotozoal Agents/toxicity , Disease Models, Animal , Drug Carriers , Epitopes , Female , Injections, Intraperitoneal , Leishmania major/immunology , Leishmaniasis Vaccines , Leishmaniasis, Cutaneous/immunology , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Nanoparticles
12.
J Neurosci ; 32(25): 8491-500, 2012 Jun 20.
Article in English | MEDLINE | ID: mdl-22723689

ABSTRACT

In neurons, the type 3 deiodinase (D3) inactivates thyroid hormone and reduces oxygen consumption, thus creating a state of cell-specific hypothyroidism. Here we show that hypoxia leads to nuclear import of D3 in neurons, without which thyroid hormone signaling and metabolism cannot be reduced. After unilateral hypoxia in the rat brain, D3 protein level is increased predominantly in the nucleus of the neurons in the pyramidal and granular ipsilateral layers, as well as in the hilus of the dentate gyrus of the hippocampal formation. In hippocampal neurons in culture as well as in a human neuroblastoma cell line (SK-N-AS), a 24 h hypoxia period redirects active D3 from the endoplasmic reticulum to the nucleus via the cochaperone Hsp40 pathway. Preventing nuclear D3 import by Hsp40 knockdown resulted an almost doubling in the thyroid hormone-dependent glycolytic rate and quadrupling the transcription of thyroid hormone target gene ENPP2. In contrast, Hsp40 overexpression increased nuclear import of D3 and minimized thyroid hormone effects in cell metabolism. In conclusion, ischemia/hypoxia induces an Hsp40-mediated translocation of D3 to the nucleus, facilitating thyroid hormone inactivation proximal to the thyroid hormone receptors. This adaptation decreases thyroid hormone signaling and may function to reduce ischemia-induced hypoxic brain damage.


Subject(s)
Cell Hypoxia/physiology , Cell Nucleus/metabolism , HSP40 Heat-Shock Proteins/physiology , Iodide Peroxidase/metabolism , Neurons/metabolism , Animals , Brain Ischemia/metabolism , Cell Nucleus/enzymology , Cells, Cultured , DNA/genetics , Endoplasmic Reticulum/metabolism , Glycosylation , Hippocampus/cytology , Hippocampus/metabolism , Immunohistochemistry , Immunoprecipitation , Male , Microscopy, Electron , Middle Cerebral Artery/physiology , Oxygen Consumption/physiology , Polymerase Chain Reaction , Rats , Rats, Sprague-Dawley , Receptors, Thyroid Hormone/metabolism , Signal Transduction/physiology , Thyroid Hormones/physiology
13.
J Neurosci Res ; 91(1): 42-50, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23073969

ABSTRACT

L1 is a cell adhesion molecule associated with a spectrum of human neurological diseases, the most well-known being X-linked hydrocephalus. L1 knockout (L1-KO) mice have revealed a variety of functions of L1 that were crucial in brain development in different brain regions. However; the function of L1 in neuronal migration during cortical histogenesis remains to be clarified. We therefore investigated the corticogenesis of mouse embryos in which L1 molecules were knocked down in selected neurons, by employing in utero electroporation with shRNAs targeting L1 (L1 shRNA). Although more than 50% of the cells transfected with no small hairpin RNA (shRNA; monster green fluorescent protein: MGFP only) vector at embryonic day 13 (E13) reached the cortical plate at E16, significantly fewer (27%) cells transfected with L1 shRNA migrated to the same extent. At E17, 22% of cells transfected with the MGFP-only vector were found in the intermediate zone, and significantly more (34%) cells transfected with L1 shRNA remained in the same zone. Furthermore, the directions of the leading process of neurons transfected with L1 shRNA became more dispersed compared with cells with the MGFP-only vector. In addition, two transcription factors expressed in the neurons, Satb2 and Tbr1, were shown to be reduced or aberrantly expressed in neurons transfected with L1 shRNA. These observations suggest that L1 plays an important role in regulating the locomotion and orientation of migrating neurons and the expression of transcription factors during neocortical development that might partially be responsible for the abnormal tract formation seen in L1-KO mice.


Subject(s)
Cell Movement/physiology , DNA-Binding Proteins/biosynthesis , Matrix Attachment Region Binding Proteins/biosynthesis , Neocortex/metabolism , Neural Cell Adhesion Molecule L1/metabolism , Neurogenesis/physiology , Transcription Factors/biosynthesis , Animals , Down-Regulation , Electroporation , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Neural Cell Adhesion Molecule L1/genetics , RNA, Small Interfering , Reverse Transcriptase Polymerase Chain Reaction , T-Box Domain Proteins
14.
Mol Cell Neurosci ; 50(2): 201-10, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22579729

ABSTRACT

Interaction of the cell adhesion molecule L1 with the cytoskeletal adaptor ankyrin is essential for topographic mapping of retinal ganglion cell (RGC) axons to synaptic targets in the superior colliculus (SC). Mice mutated in the L1 ankyrin-binding motif (FIGQY(1229)H) display abnormal mapping of RGC axons along the mediolateral axis of the SC, resembling mouse mutant phenotypes in EphB receptor tyrosine kinases. To investigate whether L1 functionally interacts with EphBs, we investigated the role of EphB kinases in phosphorylating L1 using a phospho-specific antibody to the tyrosine phosphorylated FIGQY(1229) motif. EphB2, but not an EphB2 kinase dead mutant, induced tyrosine phosphorylation of L1 at FIGQY(1229) and perturbed ankyrin recruitment to the membrane in L1-transfected HEK293 cells. Src family kinases mediated L1 phosphorylation at FIGQY(1229) by EphB2. Other EphB receptors that regulate medial-lateral retinocollicular mapping, EphB1 and EphB3, also mediated phosphorylation of L1 at FIGQY(1229). Tyrosine(1176) in the cytoplasmic domain of L1, which regulates AP2/clathrin-mediated endocytosis and axonal trafficking, was not phosphorylated by EphB2. Accordingly mutation of Tyr(1176) to Ala in L1-Y(1176)A knock-in mice resulted in normal retinocollicular mapping of ventral RGC axons. Immunostaining of the mouse SC during retinotopic mapping showed that L1 colocalized with phospho-FIGQY in RGC axons in retinorecipient layers. Immunoblotting of SC lysates confirmed that L1 was phosphorylated at FIGQY(1229) in wild type but not L1-FIGQY(1229)H (L1Y(1229)H) mutant SC, and that L1 phosphorylation was decreased in the EphB2/B3 mutant SC. Inhibition of ankyrin binding in L1Y(1229)H mutant RGCs resulted in increased neurite outgrowth compared to WT RGCs in retinal explant cultures, suggesting that L1-ankyrin binding serves to constrain RGC axon growth. These findings are consistent with a model in which EphB kinases phosphorylate L1 at FIGQY(1229) in retinal axons to modulate L1-ankyrin binding important for mediolateral retinocollicular topography.


Subject(s)
Brain Mapping , Neural Cell Adhesion Molecule L1/metabolism , Receptor, EphB2/metabolism , Retinal Ganglion Cells/physiology , Superior Colliculi/physiology , Animals , Ankyrins/metabolism , Cells, Cultured , HEK293 Cells , Humans , Mice , Mice, Mutant Strains , Mutation , Neural Cell Adhesion Molecule L1/chemistry , Neural Cell Adhesion Molecule L1/genetics , Phosphorylation , Protein Interaction Domains and Motifs , Receptor, EphB2/genetics , Receptors, Eph Family/metabolism , Retinal Ganglion Cells/metabolism , Tyrosine/genetics
15.
Mol Cell Neurosci ; 50(2): 125-35, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22561309

ABSTRACT

Injury to the central nervous system (CNS) can result in lifelong loss of function due in part to the regenerative failure of CNS neurons. Inhibitory proteins derived from myelin and the astroglial scar are major barriers for the successful regeneration of injured CNS neurons. Previously, we described the identification of a novel compound, F05, which promotes neurite growth from neurons challenged with inhibitory substrates in vitro, and promotes axonal regeneration in vivo (Usher et al., 2010). To identify additional regeneration-promoting compounds, we used F05-induced gene expression profiles to query the Broad Institute Connectivity Map, a gene expression database of cells treated with >1300 compounds. Despite no shared chemical similarity, F05-induced changes in gene expression were remarkably similar to those seen with a group of piperazine phenothiazine antipsychotics (PhAPs). In contrast to antipsychotics of other structural classes, PhAPs promoted neurite growth of CNS neurons challenged with two different glial derived inhibitory substrates. Our pharmacological studies suggest a mechanism whereby PhAPs promote growth through antagonism of calmodulin signaling, independent of dopamine receptor antagonism. These findings shed light on mechanisms underlying neurite-inhibitory signaling, and suggest that clinically approved antipsychotic compounds may be repurposed for use in CNS injured patients.


Subject(s)
Antipsychotic Agents/pharmacology , Neurites/drug effects , Phenothiazines/pharmacology , Piperazines/pharmacology , Regeneration/drug effects , Animals , Antipsychotic Agents/chemistry , Brain/physiology , CHO Cells , Calcium Signaling/drug effects , Calcium Signaling/physiology , Cell Line, Tumor , Cells, Cultured , Chondroitin Sulfate Proteoglycans/pharmacology , Cricetinae , Cricetulus , Gene Expression Profiling , Humans , Neurons/drug effects , Neurons/metabolism , Neurons/physiology , Oligonucleotide Array Sequence Analysis , Phenothiazines/chemistry , Piperazines/chemistry , Rats
16.
Mol Brain ; 16(1): 79, 2023 Nov 18.
Article in English | MEDLINE | ID: mdl-37980537

ABSTRACT

Protein kinases are responsible for protein phosphorylation and are involved in important intracellular signal transduction pathways in various cells, including neurons; however, a considerable number of poorly characterized kinases may be involved in neuronal development. Here, we considered mitogen-activated protein kinase kinase kinase kinases (MAP4Ks), related to as candidate regulators of neurite outgrowth and synaptogenesis, by examining the effects of a selective MAP4K inhibitor PF06260933. PF06260933 treatments of the cultured neurons reduced neurite lengths, not the number of synapses, and phosphorylation of GAP43 and JNK, relative to the control. These results suggest that MAP4Ks are physiologically involved in normal neuronal development and that the resultant impaired neurite outgrowth by diminished MAP4Ks' activity, is related to psychiatric disorders.


Subject(s)
Neurites , Neurons , Humans , Neurons/metabolism , Neurites/metabolism , Signal Transduction , Phosphorylation , Neuronal Outgrowth
17.
EMBO J ; 27(11): 1549-62, 2008 Jun 04.
Article in English | MEDLINE | ID: mdl-18464795

ABSTRACT

Axonal receptors for class 3 semaphorins (Sema3s) are heterocomplexes of neuropilins (Nrps) and Plexin-As signalling coreceptors. In the developing cerebral cortex, the Ig superfamily cell adhesion molecule L1 associates with Nrp1. Intriguingly, the genetic removal of L1 blocks axon responses of cortical neurons to Sema3A in vitro despite the expression of Plexin-As in the cortex, suggesting either that L1 substitutes for Plexin-As or that L1 and Plexin-A are both required and mediate distinct roles. We report that association of Nrp1 with L1 but not Plexin-As mediates the recruitment and activation of a Sema3A-induced focal adhesion kinase-mitogen-activated protein kinase cascade. This signalling downstream of L1 is needed for the disassembly of adherent points formed in growth cones and subsequently their collapse response to Sema3A. Plexin-As and L1 are coexpressed and present in common complexes in cortical neurons and both dominant-negative forms of Plexin-A and L1 impair their response to Sema3A. Consistently, Nrp1-expressing cortical projections are defective in mice lacking Plexin-A3, Plexin-A4 or L1. This reveals that specific signalling activities downstream of L1 and Plexin-As cooperate for mediating the axon guidance effects of Sema3A.


Subject(s)
Cerebral Cortex/growth & development , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Growth Cones/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neural Cell Adhesion Molecule L1/metabolism , Neuropilin-1/metabolism , Semaphorin-3A/metabolism , Animals , Axons/metabolism , Cell Adhesion , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Mice , Mice, Mutant Strains , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Cell Adhesion Molecule L1/genetics , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Signal Transduction
18.
J Cell Sci ; 123(Pt 12): 2135-43, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20501702

ABSTRACT

Hyperactivation of beta-catenin-T-cell-factor (TCF)-regulated gene transcription is a hallmark of colorectal cancer (CRC). The cell-neural adhesion molecule L1CAM (hereafter referred to as L1) is a target of beta-catenin-TCF, exclusively expressed at the CRC invasive front in humans. L1 overexpression in CRC cells increases cell growth and motility, and promotes liver metastasis. Genes induced by L1 are also expressed in human CRC tissue but the mechanisms by which L1 confers metastasis are still unknown. We found that signaling by the nuclear factor kappaB (NF-kappaB) is essential, because inhibition of signaling by the inhibitor of kappaB super repressor (IkappaB-SR) blocked L1-mediated metastasis. Overexpression of the NF-kappaB p65 subunit was sufficient to increase CRC cell proliferation, motility and metastasis. Binding of the L1 cytodomain to ezrin - a cytoskeleton-crosslinking protein - is necessary for metastasis because when binding to L1 was interrupted or ezrin gene expression was suppressed with specific shRNA, metastasis did not occur. L1 and ezrin bound to and mediated the phosphorylation of IkappaB. We also observed a complex containing IkappaB, L1 and ezrin in the juxtamembrane region of CRC cells. Furthermore, we found that L1, ezrin and phosphorylated p65 are co-expressed at the invasive front in human CRC tissue, indicating that L1-mediated activation of NF-kappaB signaling involving ezrin is a major route of CRC progression.


Subject(s)
Colorectal Neoplasms/metabolism , Cytoskeletal Proteins/metabolism , Neoplasm Metastasis , Neural Cell Adhesion Molecule L1/metabolism , Transcription Factor RelA/metabolism , Animals , Cell Line, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Cytoskeletal Proteins/genetics , Gene Expression Regulation, Neoplastic , Humans , Mice , Neural Cell Adhesion Molecule L1/genetics , Phosphorylation , Signal Transduction , Transcription Factor RelA/genetics
19.
Neuropathology ; 32(4): 420-31, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22151581

ABSTRACT

Axon regeneration is a fundamental problem facing neuroscientists and clinicians. Failure of axon regeneration is caused by both extrinsic and intrinsic mechanisms. New techniques to examine gene expression such as Next Generation Sequencing of the Transcriptome (RNA-Seq) drastically increase our knowledge of both gene expression complexity (RNA isoforms) and gene expression regulation. By utilizing RNA-Seq, gene expression can now be defined at the level of isoforms, an essential step for understanding the mechanisms governing cell identity, growth and ultimately cellular responses to injury and disease.


Subject(s)
Axons/physiology , Nerve Regeneration/physiology , Animals , Humans , Protein Isoforms/physiology
20.
Mol Cell Neurosci ; 46(1): 32-44, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20696251

ABSTRACT

Neurons in the peripheral nervous system (PNS) display a higher capacity to regenerate after injury than those in the central nervous system, suggesting cell specific transcriptional modules underlying axon growth and inhibition. We report a systems biology based search for PNS specific transcription factors (TFs). Messenger RNAs enriched in dorsal root ganglion (DRG) neurons compared to cerebellar granule neurons (CGNs) were identified using subtractive hybridization and DNA microarray approaches. Network and transcription factor binding site enrichment analyses were used to further identify TFs that may be differentially active. Combining these techniques, we identified 32 TFs likely to be enriched and/or active in the PNS. Twenty-five of these TFs were then tested for an ability to promote CNS neurite outgrowth in an overexpression screen. Real-time PCR and immunohistochemical studies confirmed that one representative TF, STAT3, is intrinsic to PNS neurons, and that constitutively active STAT3 is sufficient to promote CGN neurite outgrowth.


Subject(s)
Neurons/physiology , Peripheral Nervous System/physiology , Transcription Factors/metabolism , Transcription, Genetic , Animals , Cerebellum/cytology , Ganglia, Spinal/cytology , Gene Expression Profiling/methods , Mice , Mice, Inbred C57BL , Microarray Analysis/methods , Neurons/cytology , Nucleic Acid Hybridization/methods , RNA, Messenger/genetics , RNA, Messenger/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction/physiology , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL