Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Expert Opin Ther Targets ; 27(4-5): 269-279, 2023.
Article in English | MEDLINE | ID: mdl-37140480

ABSTRACT

INTRODUCTION: Tau has commanded much attention as a potential therapeutic target in neurodegenerative diseases. Tau pathology is a hallmark of primary tauopathies, such as progressive supranuclear palsy (PSP), corticobasal syndrome (CBS), and subtypes of frontotemporal dementia (FTD), as well as secondary tauopathies, such as Alzheimer's disease (AD). The development of tau therapeutics must reconcile with the structural complexity of the tau proteome, as well as an incomplete understanding of the role of tau in both physiology and disease. AREAS COVERED: This review offers a current perspective on tau biology, discusses key barriers to the development of effective tau-based therapeutics, and promotes the idea that pathogenic (as opposed to merely pathological) tau should be at the center of drug development efforts. EXPERT OPINION: An efficacious tau therapeutic will exhibit several primary features: 1) selectivity for pathogenic tau versus other tau species; 2) blood-brain barrier and cell membrane permeability, enabling access to intracellular tau in disease-relevant brain regions; and 3) minimal toxicity. Oligomeric tau is proposed as a major pathogenic form of tau and a compelling drug target in tauopathies.


Subject(s)
Alzheimer Disease , Supranuclear Palsy, Progressive , Tauopathies , Humans , Brain/metabolism , Supranuclear Palsy, Progressive/metabolism , Supranuclear Palsy, Progressive/pathology , tau Proteins/metabolism
2.
Proc Natl Acad Sci U S A ; 117(50): 31832-31837, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33257579

ABSTRACT

TIA1, a protein critical for eukaryotic stress response and stress granule formation, is structurally characterized in full-length form. TIA1 contains three RNA recognition motifs (RRMs) and a C-terminal low-complexity domain, sometimes referred to as a "prion-related domain" or associated with amyloid formation. Under mild conditions, full-length (fl) mouse TIA1 spontaneously oligomerizes to form a metastable colloid-like suspension. RRM2 and RRM3, known to be critical for function, are folded similarly in excised domains and this oligomeric form of apo fl TIA1, based on NMR chemical shifts. By contrast, the termini were not detected by NMR and are unlikely to be amyloid-like. We were able to assign the NMR shifts with the aid of previously assigned solution-state shifts for the RRM2,3 isolated domains and homology modeling. We present a micellar model of fl TIA1 wherein RRM2 and RRM3 are colocalized, ordered, hydrated, and available for nucleotide binding. At the same time, the termini are disordered and phase separated, reminiscent of stress granule substructure or nanoscale liquid droplets.


Subject(s)
Intrinsically Disordered Proteins/ultrastructure , T-Cell Intracellular Antigen-1/ultrastructure , Intrinsically Disordered Proteins/metabolism , Magnetic Resonance Spectroscopy , Micelles , Microscopy, Electron , Models, Molecular , Protein Folding , Protein Multimerization , RNA-Binding Motifs , Recombinant Proteins/metabolism , Recombinant Proteins/ultrastructure , T-Cell Intracellular Antigen-1/metabolism
3.
J Affect Disord ; 260: 597-603, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31541970

ABSTRACT

BACKGROUND: The TIA1 gene encodes a prion-related RNA-binding protein that regulates stress-dependent synaptic plasticity and fear memory in mice. It is unknown whether genetic variation in human TIA1 is associated with differences in stress- and fear-related behavior in people. METHODS: A longitudinal, population-based survey was conducted in Sweden to collect information on demographics, socioeconomic status, exposure to stressful life events and psychiatric symptoms. DNA samples were obtained from study participants to allow genotyping of single-nucleotide polymorphisms in the human TIA1 locus. RESULTS: We identified a single-nucleotide polymorphism in the human TIA1 gene that interacts with exposure to previous-year stressful life events to predict the development of pathological anxiety symptoms in a non-clinical cohort. LIMITATIONS: Sample population is limited in both size and scope, and we did not perform functional analysis of allelic variants of TIA1. CONCLUSIONS: TIA1 may represent a susceptibility locus for stress-dependent psychopathology. These studies support an evolutionarily conserved role of TIA1 in the mammalian brain, and may provide molecular and genetic insight into the development of stress-related psychiatric conditions such as PTSD and anxiety.


Subject(s)
Anxiety Disorders/genetics , Polymorphism, Single Nucleotide , Stress, Psychological/genetics , T-Cell Intracellular Antigen-1/genetics , Adult , Alleles , Anxiety Disorders/psychology , Cohort Studies , Female , Gene-Environment Interaction , Genotyping Techniques , Humans , Life Change Events , Longitudinal Studies , Male , Middle Aged , Risk Factors , Stress, Psychological/psychology , Sweden , Young Adult
4.
Front Behav Neurosci ; 13: 56, 2019.
Article in English | MEDLINE | ID: mdl-30967765

ABSTRACT

The generalization of fear is adaptive in that it allows an animal to respond appropriately to novel threats that are not identical to previous experiences. In contrast, the overgeneralization of fear is maladaptive and is a hallmark of post-traumatic stress disorder (PTSD), a psychiatric illness that is characterized by chronic symptomatology and a higher incidence in women compared to men. Therefore, understanding the neural basis of fear generalization at remote time-points in female animals is of particular translational relevance. However, our understanding of the neurobiology of fear generalization is largely restricted to studies employing male mice and focusing on recent time-points (i.e., within 24-48 h following conditioning). To address these limitations, we examined how male and female mice generalize contextual fear at remote time intervals (i.e., 3 weeks after conditioning). In agreement with earlier studies of fear generalization at proximal time-points, we find that the test order of training and generalization contexts is a critical determinant of generalization and context discrimination, particularly for female mice. However, tactile elements that are present during fear conditioning are more salient for male mice. Our study highlights long-term sex differences in defensive behavior between male and female mice and may provide insight into sex differences in the processing and retrieval of remote fear memory observed in humans.

5.
Cell Rep ; 26(11): 2970-2983.e4, 2019 03 12.
Article in English | MEDLINE | ID: mdl-30865887

ABSTRACT

TIA1 is a prion-related RNA-binding protein whose capacity to form various types of intracellular aggregates has been implicated in neurodegenerative disease. However, its role in normal brain function is poorly understood. Here, we show that TIA1 bidirectionally modulates stress-dependent synaptic plasticity in the hippocampus, a brain region involved in fear memory and olfactory discrimination learning. At the behavioral level, conditioned odor avoidance is potentiated by TIA1 deletion, whereas overexpression of TIA1 in the ventral hippocampus inhibits both contextual fear memory and avoidance. However, the latter genetic manipulations have little impact on other hippocampus-dependent tasks. Transcriptional profiling indicates that TIA1 presides over a large network of immune system genes with modulatory roles in synaptic plasticity and long-term memory. Our results uncover a physiological and partly sex-dependent function for TIA1 in fear memory and may provide molecular insight into stress-related psychiatric conditions, such as post-traumatic stress disorder (PTSD) and anxiety.


Subject(s)
Avoidance Learning , Fear , Memory, Long-Term , T-Cell Intracellular Antigen-1/genetics , Animals , Cytokines/genetics , Cytokines/metabolism , Female , Hippocampus/metabolism , Male , Mice , Mice, Inbred C57BL , Olfactory Perception , Sex Factors
6.
Trends Neurosci ; 42(1): 14-22, 2019 01.
Article in English | MEDLINE | ID: mdl-30391015

ABSTRACT

Over the past half-century, we have gained significant insights into the molecular biology of long-term memory storage at the level of the synapse. In recent years, our understanding of the cellular architecture supporting long-term memory traces has also substantially improved. However, the molecular biology of consolidation at the level of neuronal systems has been relatively neglected. In this opinion article, we first examine our current understanding of the cellular mechanisms of synaptic consolidation. We then outline areas requiring further investigation on how cellular changes contribute to systems consolidation. Finally, we highlight recent findings on the cellular architecture of memory traces in rodents and how the application of new technologies will expand our understanding of systems consolidation at the neural circuit level. In the coming years, this research focus will be critical for understanding the evolution of long-term memories and for enabling the development of novel therapeutics which embrace the dynamic nature of memories.


Subject(s)
Memory, Long-Term/physiology , Memory/physiology , Neuronal Plasticity/physiology , Synapses/physiology , Animals , Humans , Models, Neurological , Neurons/physiology
7.
Cell Rep ; 22(1): 59-71, 2018 01 02.
Article in English | MEDLINE | ID: mdl-29298433

ABSTRACT

Stress granules are non-membranous structures that transiently form in the cytoplasm during cellular stress, where they promote translational repression of non-essential RNAs and modulate cell signaling by sequestering key signal transduction proteins. These and other functions of stress granules facilitate an adaptive cellular response to environmental adversity. A key component of stress granules is the prion-related RNA-binding protein, T cell intracellular antigen-1 (TIA-1). Here, we report that recombinant TIA-1 undergoes rapid multimerization and phase separation in the presence of divalent zinc, which can be reversed by the zinc chelator, TPEN. Similarly, the formation and maintenance of TIA-1-positive stress granules in arsenite-treated cells are inhibited by TPEN. In addition, Zn2+ is released in cells treated with arsenite, before stress granule formation. These findings suggest that Zn2+ is a physiological ligand of TIA-1, acting as a stress-inducible second messenger to promote multimerization of TIA-1 and subsequent localization into stress granules.


Subject(s)
Arsenites/pharmacology , Cytoplasmic Granules , Protein Multimerization/drug effects , Second Messenger Systems/drug effects , T-Cell Intracellular Antigen-1 , Zinc , Cell Line , Cytoplasmic Granules/chemistry , Cytoplasmic Granules/genetics , Cytoplasmic Granules/metabolism , Humans , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , T-Cell Intracellular Antigen-1/chemistry , T-Cell Intracellular Antigen-1/genetics , T-Cell Intracellular Antigen-1/metabolism , Zinc/chemistry , Zinc/metabolism
8.
Front Behav Neurosci ; 12: 329, 2018.
Article in English | MEDLINE | ID: mdl-30697153

ABSTRACT

The generalization of fear memories is an adaptive neurobiological process that promotes survival in complex and dynamic environments. When confronted with a potential threat, an animal must select an appropriate defensive response based on previous experiences that are not identical, weighing cues and contextual information that may predict safety or danger. Like other aspects of fear memory, generalization is mediated by the coordinated actions of prefrontal, hippocampal, amygdalar, and thalamic brain areas. In this review article, we describe the current understanding of the behavioral, neural, genetic, and biochemical mechanisms involved in the generalization of fear. Fear generalization is a hallmark of many anxiety and stress-related disorders, and its emergence, severity, and manifestation are sex-dependent. Therefore, to improve the dialog between human and animal studies as well as to accelerate the development of effective therapeutics, we emphasize the need to examine both sex differences and remote timescales in rodent models.

9.
10.
Article in English | MEDLINE | ID: mdl-28049644

ABSTRACT

Prions are proteins that can adopt self-perpetuating conformations and are traditionally regarded as etiological agents of infectious neurodegenerative diseases in humans, such as Creutzfeldt-Jakob disease, kuru, and transmissible encephalopathies. More recently, a growing consensus has emerged that prion-like, self-templating mechanisms also underlie a variety of neurodegenerative disorders, including amyotrophic lateral sclerosis, Alzheimer's disease, and Huntington's disease. Perhaps most surprising, not all prion-like aggregates are associated with pathological changes. There are now several examples of prion-like proteins in mammals that serve positive biological functions in their aggregated state. In this review, we discuss functional prions in the nervous system, with particular emphasis on the cytoplasmic polyadenylation element-binding protein (CPEB) and the role of its prion-like aggregates in synaptic plasticity and memory. We also mention a more recent example of a functional prion-like protein in the brain, TIA-1, and its role during stress. These studies of functional prion-like proteins have provided a number of generalizable insights on how prion-based protein switches may operate to serve physiological functions in higher eukaryotes.


Subject(s)
Brain/metabolism , Prions/metabolism , Animals , Aplysia , Drosophila , Humans , Memory , Models, Animal , Serotonin/metabolism , Synapses/physiology , Transcription Factors/chemistry , Transcription Factors/metabolism , mRNA Cleavage and Polyadenylation Factors/chemistry , mRNA Cleavage and Polyadenylation Factors/metabolism
11.
Article in English | MEDLINE | ID: mdl-28003185

ABSTRACT

Prions are self-propagating protein conformations that are traditionally regarded as agents of neurodegenerative disease in animals. However, it has become evident that prion-like aggregation of endogenous proteins can also occur under normal physiological conditions (e.g., during memory storage or activation of the immune response). In this review, we focus on the functional prion-related protein TIA-1, an RNA-binding protein that is involved in multiple aspects of RNA metabolism but is best understood in terms of its role in stress granule assembly during the cellular stress response. We propose that stress granule formation provides a useful conceptual framework with which to address the positive role of TIA-1 prion-like aggregation. Elucidating the function of TIA-1 prion-like aggregation will advance our understanding of how prion-based molecular switches are used in normal physiological settings.


Subject(s)
Prions/metabolism , T-Cell Intracellular Antigen-1/metabolism , Animals , Humans , Memory/physiology , Neurodegenerative Diseases/physiopathology
12.
Mol Cell ; 55(2): 305-18, 2014 Jul 17.
Article in English | MEDLINE | ID: mdl-24981173

ABSTRACT

Tia1/Pub1 is a stress granule component carrying a Q/N-rich prion domain. We provide direct evidence that Tia1 forms a prion in yeast. Moreover, Tia1/Pub1 acts cooperatively with release factor Sup35/eRF3 to establish a two-protein self-propagating state. This two-protein prion driven by the Q/N-rich prion domains of Sup35 and Tia1/Pub1 can be visualized as distinctive line structures along tubulin cytoskeleton. Furthermore, we find that tubulin-associated complex containing Pub1 and Sup35 oligomers normally exists in yeast, and its assembly depends on prion domains of Pub1 and Sup35. This Sup35/Pub1 complex, which also contains TUB1 mRNA and components of translation machinery, is important for the integrity of the tubulin cytoskeleton: PUB1 disruption and Sup35 depletion from the complex lead to cytoskeletal defects. We propose that the complex is implicated in protein synthesis at the site of microtubule assembly. Thus our study identifies the role for prion domains in the assembly of multiprotein complexes.


Subject(s)
Cytoskeleton/metabolism , Peptide Termination Factors/metabolism , Poly(A)-Binding Proteins/metabolism , Prions/metabolism , RNA-Binding Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Amyloid/metabolism , Animals , Mice , Mice, Inbred C57BL , Peptide Termination Factors/chemistry , Poly(A)-Binding Proteins/chemistry , Prions/chemistry , Protein Biosynthesis , Protein Multimerization , Protein Structure, Tertiary , RNA, Fungal/metabolism , RNA, Messenger/metabolism , RNA-Binding Proteins/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , T-Cell Intracellular Antigen-1
13.
Proc Natl Acad Sci U S A ; 109(23): 9137-42, 2012 Jun 05.
Article in English | MEDLINE | ID: mdl-22619333

ABSTRACT

Whereas short-term (minutes) facilitation at Aplysia sensory-motor neuron synapses is presynaptic, long-term (days) facilitation involves synaptic growth, which requires both presynaptic and postsynaptic mechanisms. How are the postsynaptic mechanisms recruited, and when does that process begin? We have been investigating the possible role of spontaneous transmitter release from the presynaptic neuron. In the previous paper, we found that spontaneous release is critical for the induction of long-term facilitation, and this process begins during an intermediate-term stage of facilitation that is the first stage to involve postsynaptic as well as presynaptic mechanisms. We now report that increased spontaneous release during the short-term stage acts as an orthograde signal to recruit postsynaptic mechanisms of intermediate-term facilitation including increased IP3, Ca(2+), and membrane insertion and recruitment of clusters of AMPA-like receptors, which may be first steps in synaptic growth during long-term facilitation. These results suggest that the different stages of facilitation involve a cascade of pre- and postsynaptic mechanisms, which is initiated by spontaneous release and may culminate in synaptic growth.


Subject(s)
Hippocampus/metabolism , Neurotransmitter Agents/metabolism , Synaptic Transmission/physiology , Animals , Aplysia , Botulinum Toxins , Calcium/metabolism , Egtazic Acid/analogs & derivatives , Fluorescence , Hippocampus/cytology , Hygromycin B , In Situ Hybridization , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Neuronal Plasticity , Octopamine , Oligonucleotides/genetics , Organic Chemicals , Plasmids/genetics , Presynaptic Terminals/metabolism , Presynaptic Terminals/physiology , Receptors, AMPA/metabolism , Receptors, Metabotropic Glutamate/metabolism
14.
Cancer Cell ; 2(6): 463-72, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12498715

ABSTRACT

The E2F transcription factors mediate the activation or repression of key cell cycle regulatory genes under the control of the retinoblastoma protein (pRB) tumor suppressor and its relatives, p107 and p130. Here we investigate how E2F4, the major "repressive" E2F, contributes to pRB's tumor-suppressive properties. Remarkably, E2F4 loss suppresses the development of both pituitary and thyroid tumors in Rb(+/-) mice. Importantly, E2F4 loss also suppresses the inappropriate gene expression and proliferation of pRB-deficient cells. Biochemical analyses suggest that this tumor suppression occurs via a novel mechanism: E2F4 loss allows p107 and p130 to regulate the pRB-specific, activator E2Fs. We also detect these novel E2F complexes in pRB-deficient cells, suggesting that they play a significant role in the regulation of tumorigenesis in vivo.


Subject(s)
DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Proteins , Retinoblastoma Protein/deficiency , Retinoblastoma Protein/genetics , Transcription Factors/deficiency , Transcription Factors/genetics , Animals , Blotting, Western , Cell Transformation, Neoplastic/genetics , Cells, Cultured , Cyclin E/biosynthesis , DNA-Binding Proteins/metabolism , E2F4 Transcription Factor , Fibroblasts/metabolism , Mice , Mice, Mutant Strains , Mutation , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Pituitary Neoplasms/genetics , Retinoblastoma-Like Protein p107 , Retinoblastoma-Like Protein p130 , Reverse Transcriptase Polymerase Chain Reaction , Thyroid Neoplasms/genetics , Transcription Factors/metabolism
15.
Genes Dev ; 16(8): 933-47, 2002 Apr 15.
Article in English | MEDLINE | ID: mdl-11959842

ABSTRACT

Despite biochemical and genetic data suggesting that E2F and pRB (pocket protein) families regulate transcription via chromatin-modifying factors, the precise mechanisms underlying gene regulation by these protein families have not yet been defined in a physiological setting. In this study, we have investigated promoter occupancy in wild-type and pocket protein-deficient primary cells. We show that corepressor complexes consisting of histone deacetylase (HDAC1) and mSin3B were specifically recruited to endogenous E2F-regulated promoters in quiescent cells. These complexes dissociated from promoters once cells reached late G1, coincident with gene activation. Interestingly, recruitment of HDAC1 complexes to promoters depended absolutely on p107 and p130, and required an intact E2F-binding site. In contrast, mSin3B recruitment to certain promoters did not require p107 or p130, suggesting that recruitment of this corepressor can occur via E2F-dependent and -independent mechanisms. Remarkably, loss of pRB had no effect on HDAC1 or mSin3B recruitment. p107/p130 deficiency triggered a dramatic loss of E2F4 nuclear localization as well as transcriptional derepression, which is suggested by nucleosome mapping studies to be the result of localized hyperacetylation of nucleosomes proximal to E2F-binding sites. Taken together, these findings show that p130 escorts E2F4 into the nucleus and, together with corepressor complexes that contain mSin3B and/or HDAC1, directly represses transcription from target genes as cells withdraw from the cell cycle.


Subject(s)
DNA-Binding Proteins , Gene Silencing/physiology , Histone Deacetylases/metabolism , Proteins , Repressor Proteins/metabolism , Transcription Factors/metabolism , 3T3 Cells , Acetylation , Animals , Binding Sites/physiology , Cell Cycle/physiology , Cell Cycle Proteins/metabolism , Cells, Cultured , E2F Transcription Factors , E2F4 Transcription Factor , Fibroblasts/cytology , Fibroblasts/metabolism , Histone Deacetylase 1 , Macromolecular Substances , Mice , Nuclear Proteins/metabolism , Nucleosomes/metabolism , Phosphoproteins/metabolism , Promoter Regions, Genetic/physiology , Protein Binding/physiology , Retinoblastoma Protein/metabolism , Retinoblastoma-Like Protein p107 , Retinoblastoma-Like Protein p130
SELECTION OF CITATIONS
SEARCH DETAIL
...