Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Cell Genom ; 4(5): 100555, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38697121

ABSTRACT

The complex pathobiology of late-onset Alzheimer's disease (AD) poses significant challenges to therapeutic and preventative interventions. Despite these difficulties, genomics and related disciplines are allowing fundamental mechanistic insights to emerge with clarity, particularly with the introduction of high-resolution sequencing technologies. After all, the disrupted processes at the interface between DNA and gene expression, which we call the broken AD genome, offer detailed quantitative evidence unrestrained by preconceived notions about the disease. In addition to highlighting biological pathways beyond the classical pathology hallmarks, these advances have revitalized drug discovery efforts and are driving improvements in clinical tools. We review genetic, epigenomic, and gene expression findings related to AD pathogenesis and explore how their integration enables a better understanding of the multicellular imbalances contributing to this heterogeneous condition. The frontiers opening on the back of these research milestones promise a future of AD care that is both more personalized and predictive.


Subject(s)
Alzheimer Disease , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Humans , Genome, Human , Genomics/methods , Animals
2.
Sci Adv ; 9(9): eadd2671, 2023 03 03.
Article in English | MEDLINE | ID: mdl-36867706

ABSTRACT

Gene expression is changed by disease, but how these molecular responses arise and contribute to pathophysiology remains less understood. We discover that ß-amyloid, a trigger of Alzheimer's disease (AD), promotes the formation of pathological CREB3L2-ATF4 transcription factor heterodimers in neurons. Through a multilevel approach based on AD datasets and a novel chemogenetic method that resolves the genomic binding profile of dimeric transcription factors (ChIPmera), we find that CREB3L2-ATF4 activates a transcription network that interacts with roughly half of the genes differentially expressed in AD, including subsets associated with ß-amyloid and tau neuropathologies. CREB3L2-ATF4 activation drives tau hyperphosphorylation and secretion in neurons, in addition to misregulating the retromer, an endosomal complex linked to AD pathogenesis. We further provide evidence for increased heterodimer signaling in AD brain and identify dovitinib as a candidate molecule for normalizing ß-amyloid-mediated transcriptional responses. The findings overall reveal differential transcription factor dimerization as a mechanism linking disease stimuli to the development of pathogenic cellular states.


Subject(s)
Alzheimer Disease , Humans , Dimerization , Cyclic AMP Response Element-Binding Protein , Amyloid beta-Peptides , Gene Expression , Activating Transcription Factor 4 , Basic-Leucine Zipper Transcription Factors
3.
Cell Rep ; 41(3): 111488, 2022 10 18.
Article in English | MEDLINE | ID: mdl-36260999

ABSTRACT

Cells possess several conserved adaptive mechanisms to respond to stress. Stress signaling is initiated to reestablish cellular homeostasis, but its effects on the tissue or systemic levels are far less understood. We report that the secreted luminal domain of the endoplasmic reticulum (ER) stress transducer CREB3L2 (which we name TAILS [transmissible activator of increased cell livability under stress]) is an endogenous, cell non-autonomous activator of neuronal resilience. In response to oxidative insults, neurons secrete TAILS, which potentiates hedgehog signaling through direct interaction with Sonic hedgehog (SHH) and its receptor PTCH1, leading to improved antioxidant signaling and mitochondrial function in neighboring neurons. In an in vivo model of ischemic brain injury, administration of TAILS enables survival of CNS neurons and fully preserves cognitive function in behavioral tests. Our findings reveal an SHH-mediated, cell non-autonomous branch of cellular stress signaling that confers resilience to oxidative stress in the mature brain, providing protection from ischemic neurodegeneration.


Subject(s)
Antioxidants , Hedgehog Proteins , Hedgehog Proteins/metabolism , Neurons/metabolism , Oxidative Stress/physiology , Signal Transduction/physiology
4.
Cell Rep ; 29(2): 363-377.e5, 2019 Oct 08.
Article in English | MEDLINE | ID: mdl-31597097

ABSTRACT

Axon growth is regulated externally by attractive and repulsive cues generated in the environment. In addition, intrinsic pathways govern axon development, although the extent to which axons themselves can influence their own growth is unknown. We find that dorsal root ganglion (DRG) axons secrete a factor supporting axon growth and identify it as the C terminus of the ER stress-induced transcription factor CREB3L2, which is generated by site 2 protease (S2P) cleavage in sensory neurons. S2P and CREB3L2 knockdown or inhibition of axonal S2P interfere with the growth of axons, and C-terminal CREB3L2 is sufficient to rescue these effects. C-terminal CREB3L2 forms a complex with Shh and stabilizes its association with the Patched-1 receptor on developing axons. Our results reveal a neuron-intrinsic pathway downstream of S2P that promotes axon growth.


Subject(s)
Axons/metabolism , Transcription Factors/metabolism , Animals , Endopeptidases/metabolism , Ganglia, Spinal/metabolism , Gene Knockdown Techniques , HEK293 Cells , Hedgehog Proteins/metabolism , Humans , Protein Binding , Rats, Sprague-Dawley , Sensory Receptor Cells/metabolism , Signal Transduction , Transcription Factors/chemistry
5.
Neuron ; 104(5): 931-946.e5, 2019 12 04.
Article in English | MEDLINE | ID: mdl-31606248

ABSTRACT

Localized protein synthesis is fundamental for neuronal development, maintenance, and function. Transcriptomes in axons and soma are distinct, but the mechanisms governing the composition of axonal transcriptomes and their developmental regulation are only partially understood. We found that the binding motif for the RNA-binding proteins Pumilio 1 and 2 (Pum1 and Pum2) is underrepresented in transcriptomes of developing axons. Introduction of Pumilio-binding elements (PBEs) into mRNAs containing a ß-actin zipcode prevented axonal localization and translation. Pum2 is restricted to the soma of developing neurons, and Pum2 knockdown or blocking its binding to mRNA caused the appearance and translation of PBE-containing mRNAs in axons. Pum2-deficient neurons exhibited axonal growth and branching defects in vivo and impaired axon regeneration in vitro. These results reveal that Pum2 shapes axonal transcriptomes by preventing the transport of PBE-containing mRNAs into axons, and they identify somatic mRNAs retention as a mechanism for the temporal control of intra-axonal protein synthesis.


Subject(s)
Axons/metabolism , Cell Body/metabolism , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Transcriptome/physiology , Animals , Cells, Cultured , Mice , Mice, Inbred C57BL , Neurogenesis/physiology , Protein Biosynthesis/physiology , Rats , Rats, Sprague-Dawley
6.
Methods Mol Biol ; 1998: 117-128, 2019.
Article in English | MEDLINE | ID: mdl-31250298

ABSTRACT

Live imaging of microfluidically isolated axons permits study of the dynamic behavior of fluorescently tagged proteins and vesicles in these neuronal processes. We use this technique to study the motility and transport of ESCRT proteins in axons of primary hippocampal neurons. This chapter details the preparation of microfluidic chambers, as well as the seeding, fluidic isolation, and lentiviral transduction of hippocampal neurons in these chambers, optimized for the study of ESCRT protein dynamics.


Subject(s)
Axons/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism , Intravital Microscopy/methods , Microfluidic Analytical Techniques/methods , Molecular Imaging/methods , Animals , Axonal Transport , Cells, Cultured , Endosomal Sorting Complexes Required for Transport/genetics , Fluorescent Dyes/chemistry , Genetic Vectors , HEK293 Cells , Hippocampus/cytology , Humans , Lentivirus/genetics , Microfluidic Analytical Techniques/instrumentation , Molecular Probes/chemistry , Molecular Probes/genetics , Primary Cell Culture/methods , Rats , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transfection
7.
Nat Commun ; 9(1): 2189, 2018 06 06.
Article in English | MEDLINE | ID: mdl-29875359

ABSTRACT

Alternative splicing (AS) is one crucial step of gene expression that must be tightly regulated during neurodevelopment. However, the precise timing of developmental splicing switches and the underlying regulatory mechanisms are poorly understood. Here we systematically analyze the temporal regulation of AS in a large number of transcriptome profiles of developing mouse cortices, in vivo purified neuronal subtypes, and neurons differentiated in vitro. Our analysis reveals early-switch and late-switch exons in genes with distinct functions, and these switches accurately define neuronal maturation stages. Integrative modeling suggests that these switches are under direct and combinatorial regulation by distinct sets of neuronal RNA-binding proteins including Nova, Rbfox, Mbnl, and Ptbp. Surprisingly, various neuronal subtypes in the sensory systems lack Nova and/or Rbfox expression. These neurons retain the "immature" splicing program in early-switch exons, affecting numerous synaptic genes. These results provide new insights into the organization and regulation of the neurodevelopmental transcriptome.


Subject(s)
Alternative Splicing , Central Nervous System/metabolism , Gene Expression Profiling , Gene Expression Regulation, Developmental , Neurogenesis/genetics , Animals , Cell Differentiation/genetics , Central Nervous System/embryology , Central Nervous System/growth & development , Mice, Knockout , Mice, Transgenic , Models, Genetic , Models, Neurological , Neurons/cytology , Neurons/metabolism , RNA-Binding Proteins/genetics , Time Factors
8.
EMBO Rep ; 19(7)2018 07.
Article in English | MEDLINE | ID: mdl-29759981

ABSTRACT

Neurons frequently encounter neurodegenerative signals first in their periphery. For example, exposure of axons to oligomeric Aß1-42 is sufficient to induce changes in the neuronal cell body that ultimately lead to degeneration. Currently, it is unclear how the information about the neurodegenerative insult is transmitted to the soma. Here, we find that the translation of pre-localized but normally silenced sentinel mRNAs in axons is induced within minutes of Aß1-42 addition in a Ca2+-dependent manner. This immediate protein synthesis following Aß1-42 exposure generates a retrograde signaling complex including vimentin. Inhibition of the immediate protein synthesis, knock-down of axonal vimentin synthesis, or inhibition of dynein-dependent transport to the soma prevented the normal cell body response to Aß1-42 These results establish that CNS axons react to neurodegenerative insults via the local translation of sentinel mRNAs encoding components of a retrograde signaling complex that transmit the information about the event to the neuronal soma.


Subject(s)
Amyloid beta-Peptides/genetics , Nerve Degeneration/genetics , Neurons/metabolism , Peptide Fragments/genetics , RNA, Messenger/genetics , Animals , Axons/metabolism , Axons/pathology , Central Nervous System/metabolism , Dyneins/genetics , Mice , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Neurons/pathology , Rats , Signal Transduction , Transcriptome/genetics , Vimentin/genetics , Xenopus/genetics
9.
Neuron ; 97(3): 477-478, 2018 02 07.
Article in English | MEDLINE | ID: mdl-29420927

ABSTRACT

While PIWI-interacting RNAs (piRNAs) are primarily recognized as guardians of genome integrity, new functions of these small non-coding RNAs are emerging. In this issue, Kim et al. (2018) describe a piRNA-based mechanism that limits axon regeneration in C. elegans.


Subject(s)
Axons , Caenorhabditis elegans , Animals , Genome , RNA, Small Interfering , Regeneration
10.
Cell Rep ; 20(13): 3085-3098, 2017 Sep 26.
Article in English | MEDLINE | ID: mdl-28954226

ABSTRACT

Localized protein synthesis is a mechanism for developing axons to react acutely and in a spatially restricted manner to extracellular signals. As such, it is important for many aspects of axonal development, but its role in the formation of presynapses remains poorly understood. We found that the induced assembly of presynaptic terminals required local protein synthesis. Newly synthesized proteins were detectable at nascent presynapses within 15 min of inducing synapse formation in isolated axons. The transcript for the t-SNARE protein SNAP25, which is required for the fusion of synaptic vesicles with the plasma membrane, was recruited to presynaptic sites and locally translated. Inhibition of intra-axonal SNAP25 synthesis affected the clustering of SNAP25 and other presynaptic proteins and interfered with the release of synaptic vesicles from presynaptic sites. This study reveals a critical role for the axonal synthesis of SNAP25 in the assembly of presynaptic terminals.


Subject(s)
Axons/metabolism , Presynaptic Terminals/metabolism , Synaptosomal-Associated Protein 25/biosynthesis , Animals , Embryo, Mammalian , Rats , Rats, Sprague-Dawley
11.
Nat Commun ; 7: 13865, 2016 12 21.
Article in English | MEDLINE | ID: mdl-28000671

ABSTRACT

Cytoplasmic dynein mediates retrograde transport in axons, but it is unknown how its transport characteristics are regulated to meet acutely changing demands. We find that stimulus-induced retrograde transport of different cargos requires the local synthesis of different dynein cofactors. Nerve growth factor (NGF)-induced transport of large vesicles requires local synthesis of Lis1, while smaller signalling endosomes require both Lis1 and p150Glued. Lis1 synthesis is also triggered by NGF withdrawal and required for the transport of a death signal. Association of Lis1 transcripts with the microtubule plus-end tracking protein APC is required for their translation in response to NGF stimulation but not for their axonal recruitment and translation upon NGF withdrawal. These studies reveal a critical role for local synthesis of dynein cofactors for the transport of specific cargos and identify association with RNA-binding proteins as a mechanism to establish functionally distinct pools of a single transcript species in axons.


Subject(s)
Dynactin Complex/metabolism , Dyneins/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Animals , Axonal Transport/drug effects , Base Sequence , Cells, Cultured , Dynactin Complex/genetics , Female , Ganglia, Spinal/cytology , Ganglia, Spinal/embryology , Gene Expression/drug effects , Male , Nerve Growth Factor/pharmacology , Nerve Tissue Proteins/genetics , RNA Interference , Rats, Sprague-Dawley
12.
Mol Cancer Ther ; 15(4): 711-9, 2016 04.
Article in English | MEDLINE | ID: mdl-26939700

ABSTRACT

eIF4E is the key regulator of protein translation and critical for translation. The oncogenic potential of tumorigenesis, which is highly contingent on cap-dependent eIF4E, also arises from the critical role in the nuclear export and cytosolic translation of oncogenic transcripts. Inhibition of Exportin1 (XPO1), which is the major nuclear export protein for eIF4E-bound oncoprotein mRNAs, results in decreased tumor cell growth in vitro and in vivo, suggesting that eIF4E is critical in multiple myeloma. Indeed, we found that eIF4E is overexpressed in myeloma cell lines and primary myeloma cells compared with normal plasma cells. Although stable overexpression of eIF4E in multiple myeloma cells significantly increases tumorigenesis, knockdown of eIF4E impairs multiple myeloma tumor progression in a human xenograft mouse model. Using a tet-on-inducible eIF4E-knockdown system, eIF4E downregulation blocks multiple myeloma tumor growth in vivo, correlating with decreased eIF4E expression. Further overexpression and knockdown of eIF4E revealed that eIF4E regulates translation of mRNAs with highly complex 5'-untranslated regions, such as c-MYC and C/EBPß, and subsequently proliferation in multiple myeloma cells, but not in nonmalignant bone marrow stromal cells. Because many transcription factors that are critical for multiple myeloma proliferation exhibit a higher dependency on protein translation, eIF4E is an ideal and selective tool to target multiple myeloma cell growth. Mol Cancer Ther; 15(4); 711-9. ©2016 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Eukaryotic Initiation Factor-4E/metabolism , Multiple Myeloma/metabolism , Animals , Biomarkers , CCAAT-Enhancer-Binding Protein-beta/genetics , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Ectopic Gene Expression , Eukaryotic Initiation Factor-4E/antagonists & inhibitors , Eukaryotic Initiation Factor-4E/genetics , Female , Gene Expression , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Genes, myc , Humans , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , Mice , Molecular Targeted Therapy , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , RNA Interference , Signal Transduction/drug effects , Stromal Cells/drug effects , Stromal Cells/metabolism , Tumor Burden/drug effects , Tumor Burden/genetics , Xenograft Model Antitumor Assays
13.
Int J Dev Neurosci ; 55: 140-149, 2016 Dec.
Article in English | MEDLINE | ID: mdl-26970010

ABSTRACT

Proteins can be locally produced in the periphery of a cell, allowing a rapid and spatially precise response to the changes in its environment. This process is especially relevant in highly polarized and morphologically complex cells such as neurons. The study of local translation in axons has evolved from being primarily focused on developing axons, to the notion that also mature axons can produce proteins. Axonal translation has been implied in several physiological and pathological conditions, and in all cases it shares common molecular actors and pathways as well as regulatory mechanisms. Here, we review the main findings in these fields, and attempt to highlight shared principles.


Subject(s)
Axons/physiology , Nerve Regeneration/physiology , Nerve Tissue Proteins/metabolism , Neurons/physiology , Animals , Humans , Nerve Tissue Proteins/genetics , RNA, Messenger/metabolism
14.
J Vis Exp ; (100): e52805, 2015 Jun 17.
Article in English | MEDLINE | ID: mdl-26132278

ABSTRACT

Alzheimer's disease is a neurodegenerative disease affecting the aging population. A key neuropathological feature of the disease is the over-production of amyloid-beta and the deposition of amyloid-beta plaques in brain regions of the afflicted individuals. Throughout the years scientists have generated numerous Alzheimer's disease mouse models that attempt to replicate the amyloid-beta pathology. Unfortunately, the mouse models only selectively mimic the disease features. Neuronal death, a prominent effect in the brains of Alzheimer's disease patients, is noticeably lacking in these mice. Hence, we and others have employed a method of directly infusing soluble oligomeric species of amyloid-beta - forms of amyloid-beta that have been proven to be most toxic to neurons - stereotaxically into the brain. In this report we utilize male C57BL/6J mice to document this surgical technique of increasing amyloid-beta levels in a select brain region. The infusion target is the dentate gyrus of the hippocampus because this brain structure, along with the basal forebrain that is connected by the cholinergic circuit, represents one of the areas of degeneration in the disease. The results of elevating amyloid-beta in the dentate gyrus via stereotaxic infusion reveal increases in neuron loss in the dentate gyrus within 1 week, while there is a concomitant increase in cell death and cholinergic neuron loss in the vertical limb of the diagonal band of Broca of the basal forebrain. These effects are observed up to 2 weeks. Our data suggests that the current amyloid-beta infusion model provides an alternative mouse model to address region specific neuron death in a short-term basis. The advantage of this model is that amyloid-beta can be elevated in a spatial and temporal manner.


Subject(s)
Amyloid beta-Peptides/administration & dosage , Hippocampus/drug effects , Hippocampus/surgery , Neurons/drug effects , Peptide Fragments/administration & dosage , Animals , Hippocampus/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Stereotaxic Techniques
15.
J Vis Exp ; (100): e52799, 2015 Jun 17.
Article in English | MEDLINE | ID: mdl-26131922

ABSTRACT

mRNAs are frequently localized to vertebrate axons and their local translation is required for axon pathfinding or branching during development and for maintenance, repair or neurodegeneration in postdevelopmental periods. High throughput analyses have recently revealed that axons have a more dynamic and complex transcriptome than previously expected. These analysis, however have been mostly done in cultured neurons where axons can be isolated from the somato-dendritic compartments. It is virtually impossible to achieve such isolation in whole tissues in vivo. Thus, in order to verify the recruitment of mRNAs and their functional relevance in a whole animal, transcriptome analyses should ideally be combined with techniques that allow the visualization of mRNAs in situ. Recently, novel ISH technologies that detect RNAs at a single-molecule level have been developed. This is especially important when analyzing the subcellular localization of mRNA, since localized RNAs are typically found at low levels. Here we describe two protocols for the detection of axonally-localized mRNAs using a novel ultrasensitive RNA ISH technology. We have combined RNAscope ISH with axonal counterstain using fluorescence immunohistochemistry or histological dyes to verify the recruitment of Atf4 mRNA to axons in vivo in the mature mouse and human brains.


Subject(s)
Axons/chemistry , Brain Chemistry , In Situ Hybridization/methods , RNA, Messenger/analysis , Activating Transcription Factor 4/genetics , Animals , Brain/metabolism , Humans , Immunohistochemistry , Mice
16.
Nat Commun ; 6: 6888, 2015 Apr 22.
Article in English | MEDLINE | ID: mdl-25901863

ABSTRACT

Local translation mediates axonal responses to Semaphorin3A (Sema3A) and other guidance cues. However, only a subset of the axonal proteome is locally synthesized, whereas most proteins are trafficked from the soma. The reason why only specific proteins are locally synthesized is unknown. Here we show that local protein synthesis and degradation are linked events in growth cones. We find that growth cones exhibit high levels of ubiquitination and that local signalling pathways trigger the ubiquitination and degradation of RhoA, a mediator of Sema3A-induced growth cone collapse. Inhibition of RhoA degradation is sufficient to remove the protein-synthesis requirement for Sema3A-induced growth cone collapse. In addition to RhoA, we find that locally translated proteins are the main targets of the ubiquitin-proteasome system in growth cones. Thus, local protein degradation is a major feature of growth cones and creates a requirement for local translation to replenish proteins needed to maintain growth cone responses.


Subject(s)
Growth Cones/metabolism , Neurons/metabolism , RNA, Messenger/metabolism , Ubiquitin-Protein Ligases/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Axons/drug effects , Axons/metabolism , Blotting, Western , Electroporation , Embryo, Mammalian , Ganglia, Spinal/cytology , Growth Cones/drug effects , HEK293 Cells , Hippocampus/cytology , Humans , Mice , Nerve Growth Factor/pharmacology , Neurons/cytology , Neurons/drug effects , PC12 Cells , Proteasome Endopeptidase Complex , Protein Biosynthesis , Rats , Rats, Sprague-Dawley , Semaphorin-3A/pharmacology , Spinal Cord , Ubiquitin-Protein Ligases/drug effects , Ubiquitination/drug effects
17.
Neurotherapeutics ; 12(1): 57-65, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25371166

ABSTRACT

Localized protein synthesis is a mechanism by which morphologically polarized cells react in a spatially confined and temporally acute manner to changes in their environment. During the development of the nervous system intra-axonal protein synthesis is crucial for the establishment of neuronal connections. In contrast, mature axons have long been considered as translationally inactive but upon nerve injury or under neurodegenerative conditions specific subsets of mRNAs are recruited into axons and locally translated. Intra-axonally synthesized proteins can have pathogenic or restorative and regenerative functions, and thus targeting the axonal translatome might have therapeutic value, for example in the treatment of spinal cord injury or Alzheimer's disease. In the case of Alzheimer's disease the local synthesis of the stress response transcription factor activating transcription factor 4 mediates the long-range retrograde spread of pathology across the brain, and inhibition of local Atf4 translation downstream of the integrated stress response might interfere with this spread. Several molecular tools and approaches have been developed to target specifically the axonal translatome by either overexposing proteins locally in axons or, conversely, knocking down selectively axonally localized mRNAs. Many questions about axonal translation remain to be answered, especially with regard to the mechanisms establishing specificity but, nevertheless, targeting the axonal translatome is a promising novel avenue to pursue in the development for future therapies for various neurological conditions.


Subject(s)
Axons/metabolism , Nerve Degeneration/metabolism , Nerve Regeneration/physiology , Protein Biosynthesis/physiology , Animals , Humans , Neurodegenerative Diseases/metabolism
18.
Cell ; 158(5): 1159-1172, 2014 Aug 28.
Article in English | MEDLINE | ID: mdl-25171414

ABSTRACT

In Alzheimer's disease (AD) brain, exposure of axons to Aß causes pathogenic changes that spread retrogradely by unknown mechanisms, affecting the entire neuron. We found that locally applied Aß1-42 initiates axonal synthesis of a defined set of proteins including the transcription factor ATF4. Inhibition of local translation and retrograde transport or knockdown of axonal Atf4 mRNA abolished Aß-induced ATF4 transcriptional activity and cell loss. Aß1-42 injection into the dentate gyrus (DG) of mice caused loss of forebrain neurons whose axons project to the DG. Protein synthesis and Atf4 mRNA were upregulated in these axons, and coinjection of Atf4 siRNA into the DG reduced the effects of Aß1-42 in the forebrain. ATF4 protein and transcripts were found with greater frequency in axons in the brain of AD patients. These results reveal an active role for intra-axonal translation in neurodegeneration and identify ATF4 as a mediator for the spread of AD pathology.


Subject(s)
Activating Transcription Factor 4/analysis , Alzheimer Disease/pathology , Brain/pathology , Activating Transcription Factor 4/metabolism , Amyloid beta-Peptides/genetics , Animals , Axons/metabolism , Brain/cytology , Brain Chemistry , Eukaryotic Initiation Factor-2/metabolism , Hippocampus , Humans , Mice, Inbred C57BL , Rats , Transcription Factor CHOP/metabolism
19.
Nat Commun ; 5: 3506, 2014 Mar 25.
Article in English | MEDLINE | ID: mdl-24667291

ABSTRACT

The surface of developing axons expands in a process mediated by the exocyst complex. The spatio-temporal regulation of the exocyst is only partially understood. Here we report that stimulated membrane enlargement in dorsal root ganglion (DRG) axons is triggered by intra-axonal synthesis of TC10, a small GTPase required for exocyst function. Induced membrane expansion and axon outgrowth are inhibited after axon-specific knockdown of TC10 mRNA. To determine the relationship of intra-axonal TC10 synthesis with the previously described stimulus-induced translation of the cytoskeletal regulator Par3, we investigate the signalling pathways controlling their local translation in response to NGF. Phosphoinositide 3-kinase (PI3K)-dependent activation of the Rheb-mTOR pathway triggers the simultaneous local synthesis of TC10 and Par3. These results reveal the importance of local translation in the control of membrane dynamics and demonstrate that localized, mTOR-dependent protein synthesis triggers the simultaneous activation of parallel pathways.


Subject(s)
Axons/metabolism , Carrier Proteins/genetics , Ganglia, Spinal/metabolism , Neurons/metabolism , RNA, Messenger/metabolism , rho GTP-Binding Proteins/genetics , Animals , Axons/drug effects , Carrier Proteins/drug effects , Carrier Proteins/metabolism , Cells, Cultured , Ganglia, Spinal/cytology , Ganglia, Spinal/drug effects , Monomeric GTP-Binding Proteins/drug effects , Monomeric GTP-Binding Proteins/metabolism , Nerve Growth Factor/pharmacology , Nerve Tissue Proteins , Neurons/cytology , Neurons/drug effects , Neuropeptides/drug effects , Neuropeptides/metabolism , Phosphatidylinositol 3-Kinase/drug effects , Phosphatidylinositol 3-Kinase/metabolism , RNA, Messenger/drug effects , Ras Homolog Enriched in Brain Protein , Rats , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/drug effects , TOR Serine-Threonine Kinases/metabolism , rho GTP-Binding Proteins/drug effects , rho GTP-Binding Proteins/metabolism
20.
Nat Cell Biol ; 11(8): 1024-30, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19620967

ABSTRACT

During development, axon growth rates are precisely regulated to provide temporal control over pathfinding. The precise temporal regulation of axonal growth is a key step in the formation of functional synapses and the proper patterning of the nervous system. The rate of axonal elongation is increased by factors such as netrin-1 and nerve growth factor (NGF), which stimulate axon outgrowth using incompletely defined pathways. To clarify the mechanism of netrin-1- and NGF-stimulated axon growth, we explored the role of local protein translation. We found that intra-axonal protein translation is required for stimulated, but not basal, axon outgrowth. To identify the mechanism of translation-dependent outgrowth, we examined the PAR complex, a cytoskeleton regulator. We found that the PAR complex, like local translation, is required for stimulated, but not basal, outgrowth. Par3 mRNA is localized to developing axons, and NGF and netrin-1 trigger its local translation. Selective ablation of Par3 mRNA from axons abolishes the outgrowth-promoting effect of NGF. These results identify a new role for local translation and the PAR complex in axonal outgrowth.


Subject(s)
Axons/drug effects , Nerve Growth Factor/pharmacology , Nerve Growth Factors/pharmacology , Protein Biosynthesis/drug effects , Tumor Suppressor Proteins/pharmacology , Adaptor Proteins, Signal Transducing , Animals , Axons/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Culture Techniques/instrumentation , Cell Polarity , Cells, Cultured , Ganglia, Spinal/cytology , Ganglia, Spinal/embryology , Gene Expression/drug effects , In Situ Hybridization, Fluorescence , Microscopy, Fluorescence , Nerve Tissue Proteins , Netrin-1 , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Protein Binding , Protein Kinase C/genetics , Protein Kinase C/metabolism , RNA Interference , Rats , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...