Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters










Publication year range
1.
Nat Aging ; 4(6): 791-813, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38750187

ABSTRACT

Classical evolutionary theories propose tradeoffs among reproduction, damage repair and lifespan. However, the specific role of the germline in shaping vertebrate aging remains largely unknown. In this study, we used the turquoise killifish (Nothobranchius furzeri) to genetically arrest germline development at discrete stages and examine how different modes of infertility impact life history. We first constructed a comprehensive single-cell gonadal atlas, providing cell-type-specific markers for downstream phenotypic analysis. We show here that germline depletion-but not arresting germline differentiation-enhances damage repair in female killifish. Conversely, germline-depleted males instead showed an extension in lifespan and rejuvenated metabolic functions. Through further transcriptomic analysis, we highlight enrichment of pro-longevity pathways and genes in germline-depleted male killifish and demonstrate functional conservation of how these factors may regulate longevity in germline-depleted Caenorhabditis elegans. Our results, therefore, demonstrate that different germline manipulation paradigms can yield pronounced sexually dimorphic phenotypes, implying alternative responses to classical evolutionary tradeoffs.


Subject(s)
Germ Cells , Longevity , Animals , Longevity/genetics , Male , Female , Germ Cells/metabolism , Caenorhabditis elegans/genetics , Caenorhabditis elegans/physiology , Sex Characteristics
2.
bioRxiv ; 2024 May 12.
Article in English | MEDLINE | ID: mdl-38187630

ABSTRACT

Classical evolutionary theories propose tradeoffs between reproduction, damage repair, and lifespan. However, the specific role of the germline in shaping vertebrate aging remains largely unknown. Here, we use the turquoise killifish ( N. furzeri ) to genetically arrest germline development at discrete stages, and examine how different modes of infertility impact life-history. We first construct a comprehensive single-cell gonadal atlas, providing cell-type-specific markers for downstream phenotypic analysis. Next, we show that germline depletion - but not arresting germline differentiation - enhances damage repair in female killifish. Conversely, germline-depleted males instead showed an extension in lifespan and rejuvenated metabolic functions. Through further transcriptomic analysis, we highlight enrichment of pro-longevity pathways and genes in germline-depleted male killifish and demonstrate functional conservation of how these factors may regulate longevity in germline-depleted C. elegans . Our results therefore demonstrate that different germline manipulation paradigms can yield pronounced sexually dimorphic phenotypes, implying alternative responses to classical evolutionary tradeoffs.

3.
Front Mol Biosci ; 10: 1290118, 2023.
Article in English | MEDLINE | ID: mdl-38016061

ABSTRACT

The protein homeostasis (proteostasis) network is a nexus of molecular mechanisms that act in concert to maintain the integrity of the proteome and ensure proper cellular and organismal functionality. Early in life the proteostasis network efficiently preserves the functionality of the proteome, however, as the organism ages, or due to mutations or environmental insults, subsets of inherently unstable proteins misfold and form insoluble aggregates that accrue within the cell. These aberrant protein aggregates jeopardize cellular viability and, in some cases, underlie the development of devastating illnesses. Hence, the accumulation of protein aggregates activates different nodes of the proteostasis network that refold aberrantly folded polypeptides, or direct them for degradation. The proteostasis network apparently functions within the cell, however, a myriad of studies indicate that this nexus of mechanisms is regulated at the organismal level by signaling pathways. It was also discovered that the proteostasis network differentially responds to dissimilar proteotoxic insults by tailoring its response according to the specific challenge that cells encounter. In this mini-review, we delineate the proteostasis-regulating neuronal mechanisms, describe the indications that the proteostasis network differentially responds to distinct proteotoxic challenges, and highlight possible future clinical prospects of these insights.

4.
Aging Cell ; 22(12): e14013, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37897137

ABSTRACT

Aberrant protein aggregation jeopardizes cellular functionality and underlies the development of a myriad of late-onset maladies including Alzheimer's disease (AD) and Huntington's disease (HD). Accordingly, molecules that mitigate the toxicity of hazardous protein aggregates are of great interest as potential future therapeutics. Here we asked whether a small peptide, composed of five amino acids (5MER peptide) that was derived from the human pro-inflammatory CD44 protein, could protect model nematodes from the toxicity of aggregative proteins that underlie the development of neurodegenerative disorders in humans. We found that the 5MER peptide mitigates the toxicity that stems from both; the AD-causing Aß peptide and a stretch of poly-glutamine that is accountable for the development of several disorders including HD, while minimally affecting lifespan. This protection was dependent on the activity of aging-regulating transcription factors and associated with enhanced Aß and polyQ35-YFP aggregation. A transcriptomic analysis unveiled that the peptide modifies signaling pathways, thereby modulating the expression of various genes, including these, which are known as protein homeostasis (proteostasis) regulators such as txt-13 and modifiers of proteasome activity. The knockdown of txt-13 protects worms from proteotoxicity to the same extent as the 5MER peptide, suggesting that the peptide activates the transcellular chaperone signaling to promote proteostasis. Together, our results propose that the 5MER peptide should be considered as a component of future therapeutic cocktails for the treatment of neurodegenerative maladies.


Subject(s)
Alzheimer Disease , Caenorhabditis elegans , Animals , Humans , Caenorhabditis elegans/genetics , Amyloid beta-Peptides/toxicity , Amyloid beta-Peptides/metabolism , Transcription Factors/metabolism , Alzheimer Disease/drug therapy , Aging
5.
Cell Rep ; 38(6): 110350, 2022 02 08.
Article in English | MEDLINE | ID: mdl-35139369

ABSTRACT

The protein homeostasis (proteostasis) network (PN) encompasses mechanisms that maintain proteome integrity by controlling various biological functions. Loss of proteostasis leads to toxic protein aggregation (proteotoxicity), which underlies the manifestation of neurodegeneration. How the PN responds to dissimilar proteotoxic challenges and how these responses are regulated at the organismal level are largely unknown. Here, we report that, while torsin chaperones protect from the toxicity of neurodegeneration-causing polyglutamine stretches, they exacerbate the toxicity of the Alzheimer's disease-causing Aß peptide in neurons and muscles. These opposing effects are accompanied by differential modulations of gene expression, including that of three neuropeptides that are involved in tailoring the organismal response to dissimilar proteotoxic insults. This mechanism is regulated by insulin/IGF signaling and the transcription factor SKN-1/NRF. Our work delineates a mechanism by which the PN orchestrates differential responses to dissimilar proteotoxic challenges and points at potential targets for therapeutic interventions.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , DNA-Binding Proteins/metabolism , Neuropeptides/metabolism , Proteostasis/physiology , Signal Transduction/physiology , Transcription Factors/metabolism , Animals , Caenorhabditis elegans/metabolism , Homeostasis/physiology , Peptides/metabolism , Proteome/metabolism
6.
PLoS One ; 16(7): e0243522, 2021.
Article in English | MEDLINE | ID: mdl-34197476

ABSTRACT

Lowering the activity of the Insulin/IGF-1 Signaling (IIS) cascade results in elevated stress resistance, enhanced protein homeostasis (proteostasis) and extended lifespan of worms, flies and mice. In the nematode Caenorhabditis elegans (C. elegans), the longevity phenotype that stems from IIS reduction is entirely dependent upon the activities of a subset of transcription factors including the Forkhead factor DAF-16/FOXO (DAF-16), Heat Shock Factor-1 (HSF-1), SKiNhead/Nrf (SKN-1) and ParaQuat Methylviologen responsive (PQM-1). While DAF-16 determines lifespan exclusively during early adulthood and governs proteostasis in early adulthood and midlife, HSF-1 executes these functions foremost during development. Despite the central roles of SKN-1 as a regulator of lifespan and proteostasis, the temporal requirements of this transcription factor were unknown. Here we employed conditional knockdown techniques and discovered that in C. elegans, SKN-1 is primarily important for longevity and proteostasis during late larval development through early adulthood. Our findings indicate that events that occur during late larval developmental through early adulthood affect lifespan and proteostasis and suggest that subsequent to HSF-1, SKN-1 sets the conditions, partially overlapping temporally with DAF-16, that enable IIS reduction to promote longevity and proteostasis. Our findings raise the intriguing possibility that HSF-1, SKN-1 and DAF-16 function in a coordinated and sequential manner to promote healthy aging.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/physiology , DNA-Binding Proteins/metabolism , Longevity , Proteostasis/physiology , Transcription Factors/metabolism , Animals , Caenorhabditis elegans/growth & development , Caenorhabditis elegans Proteins/antagonists & inhibitors , Caenorhabditis elegans Proteins/genetics , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Forkhead Transcription Factors/metabolism , Larva/drug effects , Larva/growth & development , Larva/metabolism , Peptides/pharmacology , RNA Interference , RNA, Double-Stranded/metabolism , Ribonuclease III/antagonists & inhibitors , Ribonuclease III/genetics , Ribonuclease III/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics
7.
Genetics ; 215(4): 889-901, 2020 08.
Article in English | MEDLINE | ID: mdl-32759342

ABSTRACT

Sustaining a healthy proteome is a lifelong challenge for each individual cell of an organism. However, protein homeostasis or proteostasis is constantly jeopardized since damaged proteins accumulate under proteotoxic stress that originates from ever-changing metabolic, environmental, and pathological conditions. Proteostasis is achieved via a conserved network of quality control pathways that orchestrate the biogenesis of correctly folded proteins, prevent proteins from misfolding, and remove potentially harmful proteins by selective degradation. Nevertheless, the proteostasis network has a limited capacity and its collapse deteriorates cellular functionality and organismal viability, causing metabolic, oncological, or neurodegenerative disorders. While cell-autonomous quality control mechanisms have been described intensely, recent work on Caenorhabditis elegans has demonstrated the systemic coordination of proteostasis between distinct tissues of an organism. These findings indicate the existence of intricately balanced proteostasis networks important for integration and maintenance of the organismal proteome, opening a new door to define novel therapeutic targets for protein aggregation diseases. Here, we provide an overview of individual protein quality control pathways and the systemic coordination between central proteostatic nodes. We further provide insights into the dynamic regulation of cellular and organismal proteostasis mechanisms that integrate environmental and metabolic changes. The use of C. elegans as a model has pioneered our understanding of conserved quality control mechanisms important to safeguard the organismal proteome in health and disease.


Subject(s)
Caenorhabditis elegans/physiology , HSP70 Heat-Shock Proteins/metabolism , Homeostasis , Proteome/metabolism , Proteostasis Deficiencies/physiopathology , Proteostasis , Stress, Physiological , Animals , Humans , Protein Folding , Signal Transduction
8.
Biomolecules ; 10(4)2020 03 26.
Article in English | MEDLINE | ID: mdl-32225014

ABSTRACT

Despite huge investments and major efforts to develop remedies for Alzheimer's disease (AD) in the past decades, AD remains incurable. While evidence for molecular and phenotypic variability in AD have been accumulating, AD research still heavily relies on the search for AD-specific genetic/protein biomarkers that are expected to exhibit repetitive patterns throughout all patients. Thus, the classification of AD patients to different categories is expected to set the basis for the development of therapies that will be beneficial for subpopulations of patients. Here we explore the molecular heterogeneity among a large cohort of AD and non-demented brain samples, aiming to address the question whether AD-specific molecular biomarkers can progress our understanding of the disease and advance the development of anti-AD therapeutics. We studied 951 brain samples, obtained from up to 17 brain regions of 85 AD patients and 22 non-demented subjects. Utilizing an information-theoretic approach, we deciphered the brain sample-specific structures of altered transcriptional networks. Our in-depth analysis revealed that 7 subnetworks were repetitive in the 737 diseased and 214 non-demented brain samples. Each sample was characterized by a subset consisting of ~1-3 subnetworks out of 7, generating 52 distinct altered transcriptional signatures that characterized the 951 samples. We show that 30 different altered transcriptional signatures characterized solely AD samples and were not found in any of the non-demented samples. In contrast, the rest of the signatures characterized different subsets of sample types, demonstrating the high molecular variability and complexity of gene expression in AD. Importantly, different AD patients exhibiting similar expression levels of AD biomarkers harbored distinct altered transcriptional networks. Our results emphasize the need to expand the biomarker-based stratification to patient-specific transcriptional signature identification for improved AD diagnosis and for the development of subclass-specific future treatment.


Subject(s)
Alzheimer Disease/genetics , Brain/physiopathology , Transcriptome/genetics , Aged , Aged, 80 and over , Alzheimer Disease/physiopathology , Apolipoproteins E/genetics , Calcium Signaling/genetics , Case-Control Studies , Databases, Genetic , Humans , Middle Aged , Precision Medicine/methods , Reproducibility of Results
9.
Trends Cell Biol ; 29(12): 954-963, 2019 12.
Article in English | MEDLINE | ID: mdl-31669295

ABSTRACT

The proteostasis network (PN) is a nexus of mechanisms that act in concert to maintain the integrity of the proteome. Efficiency of the PN declines with age, resulting in the accumulation of misfolded proteins, and in some cases in the development of neurodegenerative disorders. Thus, maintaining an active and efficient PN through the late stages of life could delay or prevent neurodegeneration. Indeed, altering the activity of aging-regulating pathways protects model organisms from neurodegeneration-linked toxic protein aggregation. Here, we delineate evidence that the formation and integrity of lipid assemblies are affected by aging-regulating pathways, and describe the roles of these structures in proteostasis maintenance. We also highlight future research directions and discuss the possibility that compounds which modulate lipid assemblies could be used for the treatment of neurodegenerative disorders.


Subject(s)
Aging/physiology , Protein Folding , Proteostasis/physiology , Animals , Humans , Lipids , Neurodegenerative Diseases/pathology , Proteome/metabolism , Proteostasis Deficiencies/pathology
10.
Aging Cell ; 18(6): e13047, 2019 12.
Article in English | MEDLINE | ID: mdl-31576648

ABSTRACT

Cellular mechanisms that act in concert to maintain protein homeostasis (proteostasis) are vital for organismal functionality and survival. Nevertheless, subsets of aggregation-prone proteins form toxic aggregates (proteotoxicity) that in some cases, underlie the development of neurodegenerative diseases. Proteotoxic aggregates are often deposited in the vicinity of the nucleus, a process that is cytoskeleton-dependent. Accordingly, cytoskeletal dysfunction contributes to pathological hallmarks of various neurodegenerative diseases. Here, we asked whether the linker of nucleoskeleton and cytoskeleton (LINC) complex, which bridges these filaments across the nuclear envelope, is needed for the maintenance of proteostasis. Employing model nematodes, we discovered that knocking down LINC components impairs the ability of the worm to cope with proteotoxicity. Knocking down anc-1, which encodes a key component of the LINC complex, modulates the expression of transcription factors and E3 ubiquitin ligases, thereby affecting the rates of protein ubiquitination and impairing proteasome-mediated protein degradation. Our results establish a link between the LINC complex, protein degradation, and neurodegeneration-associated proteotoxicity.


Subject(s)
Caenorhabditis elegans/genetics , Cytoskeleton/genetics , Gene Expression Regulation , Nuclear Matrix/genetics , Proteasome Endopeptidase Complex/genetics , Proteostasis/genetics , Animals , Caenorhabditis elegans/metabolism , Cytoskeleton/metabolism , Gene Expression Profiling , Nuclear Matrix/metabolism , Proteasome Endopeptidase Complex/metabolism , Sequence Analysis, RNA
11.
J Mol Biol ; 431(9): 1711-1728, 2019 04 19.
Article in English | MEDLINE | ID: mdl-30878478

ABSTRACT

Myotonic dystrophy type 1 is an autosomal-dominant inherited disorder caused by the expansion of CTG repeats in the 3' untranslated region of the DMPK gene. The RNAs bearing these expanded repeats have a range of toxic effects. Here we provide evidence from a Caenorhabditis elegans myotonic dystrophy type 1 model that the RNA interference (RNAi) machinery plays a key role in causing RNA toxicity and disease phenotypes. We show that the expanded repeats systematically affect a range of endogenous genes bearing short non-pathogenic repeats and that this mechanism is dependent on the small RNA pathway. Conversely, by perturbating the RNA interference machinery, we reversed the RNA toxicity effect and reduced the disease pathogenesis. Our results unveil a role for RNA repeats as templates (based on sequence homology) for moderate but constant gene silencing. Such a silencing effect affects the cell steady state over time, with diverse impacts depending on tissue, developmental stage, and the type of repeat. Importantly, such a mechanism may be common among repeats and similar in human cells with different expanded repeat diseases.


Subject(s)
Aging/genetics , Caenorhabditis elegans/genetics , Myotonic Dystrophy/genetics , RNA Interference , RNA, Double-Stranded/genetics , Trinucleotide Repeats , 3' Untranslated Regions , Animals , Animals, Genetically Modified , Caenorhabditis elegans/growth & development , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Disease Models, Animal , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Hot Temperature , Humans , Myotonic Dystrophy/metabolism , Myotonic Dystrophy/pathology , Protein Binding , RNA, Double-Stranded/metabolism , RNA, Helminth/genetics , RNA, Helminth/metabolism
12.
Elife ; 72018 11 07.
Article in English | MEDLINE | ID: mdl-30403374

ABSTRACT

Although aging-regulating pathways were discovered a few decades ago, it is not entirely clear how their activities are orchestrated, to govern lifespan and proteostasis at the organismal level. Here, we utilized the nematode Caenorhabditis elegans to examine whether the alteration of aging, by reducing the activity of the Insulin/IGF signaling (IIS) cascade, affects protein SUMOylation. We found that IIS activity promotes the SUMOylation of the germline protein, CAR-1, thereby shortening lifespan and impairing proteostasis. In contrast, the expression of mutated CAR-1, that cannot be SUMOylated at residue 185, extends lifespan and enhances proteostasis. A mechanistic analysis indicated that CAR-1 mediates its aging-altering functions, at least partially, through the notch-like receptor glp-1. Our findings unveil a novel regulatory axis in which SUMOylation is utilized to integrate the aging-controlling functions of the IIS and of the germline and provide new insights into the roles of SUMOylation in the regulation of organismal aging.


Subject(s)
Aging/metabolism , Caenorhabditis elegans/metabolism , Insulin-Like Growth Factor I/metabolism , Insulin/metabolism , Proteostasis , Signal Transduction , Sumoylation , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/metabolism , Gene Knockdown Techniques , Germ Cells/metabolism , Gonads/metabolism , Longevity , Models, Biological , Stress, Physiological , Transcription, Genetic
13.
EMBO Rep ; 19(8)2018 08.
Article in English | MEDLINE | ID: mdl-29945933

ABSTRACT

Reducing insulin/IGF-1 signaling (IIS) extends lifespan, promotes protein homeostasis (proteostasis), and elevates stress resistance of worms, flies, and mammals. How these functions are orchestrated across the organism is only partially understood. Here, we report that in the nematode Caenorhabditis elegans, the IIS positively regulates the expression of caveolin-1 (cav-1), a gene which is primarily expressed in neurons of the adult worm and underlies the formation of caveolae, a subtype of lipid microdomains that serve as platforms for signaling complexes. Accordingly, IIS reduction lowers cav-1 expression and lessens the quantity of neuronal caveolae. Reduced cav-1 expression extends lifespan and mitigates toxic protein aggregation by modulating the expression of aging-regulating and signaling-promoting genes. Our findings define caveolae as aging-governing signaling centers and underscore the potential for cav-1 as a novel therapeutic target for the promotion of healthy aging.


Subject(s)
Aging/metabolism , Caenorhabditis elegans/metabolism , Caveolae/metabolism , Insulin-Like Growth Factor I/metabolism , Insulin/metabolism , Signal Transduction , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/ultrastructure , Caenorhabditis elegans Proteins/metabolism , Caveolae/ultrastructure , Caveolin 1/metabolism , Caveolin 2/metabolism , Gene Expression Regulation , Gene Knockdown Techniques , Heat-Shock Response , Longevity , Models, Biological , Proteostasis , RNA Interference , Transcription Factors/metabolism , Ultraviolet Rays
14.
J Mater Chem B ; 6(48): 8228-8237, 2018 Dec 28.
Article in English | MEDLINE | ID: mdl-32254943

ABSTRACT

Self-assembly of biomolecules facilitates the formation of a diverse range of nanostructures from a wide range of materials. Peptides, specifically short peptides, are very useful in this respect due to their biocompatibility, ease of synthesis, functionality and tunable bioactivity. As a result, understanding the factors that rule the morphology of the self assembled nanostructures is extremely important. Furthermore, the applications of these self-assembled nanostructures in biomedical research have intrigued researchers for a long time and recently witnessed an exponential growth. Here, we report the design and synthesis of two short (tri) peptides with similar backbones and their corresponding Cu(ii) conjugates. Variation in the hydrophobicity of the central amino acid in the peptide backbone and the introduction of a metal-peptide coordination center rule the self assembly process in such a fashion that it generates various nanostructures with different morphologies. More importantly, these metallo-peptide assemblies can serve as a simple and spontaneous drug delivery system. The system delivers the drug using a fluorescence-based displacement strategy with a turn-on emission response. The naturally occurring amino acid, histidine, displaces and releases the metallo-peptide-bound drug in a controlled and immediate manner. We demonstrated the activity of this system using the efficient anticancer chemotherapy drug doxorubicin (DOX). This strategy parallelly allows the release as well as the trace of the location of the drug. Moreover, we confirmed that the system is not cytotoxic and has high cellular stability. To the best of our knowledge, this is the first report on the use of metallo-peptides as an optical-based drug displacement system.

15.
FASEB J ; 32(3): 1479-1492, 2018 03.
Article in English | MEDLINE | ID: mdl-29127190

ABSTRACT

Loss of protein homeostasis is a hazardous situation that jeopardizes cellular functionality and viability. Cells have developed mechanisms that supervise protein integrity and direct misfolded molecules for degradation. Nevertheless, subsets of aggregation-prone proteins escape degradation and form aggregates that can underlie the development of neurodegenerative disorders. In some cases, cells deposit hazardous protein aggregates in designated sites, like aggresomes, or secrete them with vesicles. The prion protein (PrP) is an aggregation-prone, membrane-anchored glycoprotein, whose aggregation causes familial and sporadic, fatal, neurodegenerative diseases. The proper maturation of PrP is assisted by cyclophilin B, an endoplasmic reticulum-resident foldase. Accordingly, the inhibition of cyclophilins by the drug cyclosporin A (CsA) leads to the accumulation of aggregated PrP and to its deposition in aggresomes. In this study, we asked whether secretion is an alternative strategy that cells adopt to get rid of misfolded PrP molecules and found that, upon treatment with CsA, cells secrete PrP by exosomes, a subtype of secretion vesicles, and by additional types of vesicles. CsA-induced, PrP-containing exosomes originate from the endoplasmic reticulum in a Golgi-independent manner. These findings divulge a new cellular response that is activated upon CsA treatment to secrete misfolded PrP species from the cell and may underlie the spreading of toxic prions among cells and across tissues.-Pan, I., Roitenberg, N., Cohen, E. Vesicle-mediated secretion of misfolded prion protein molecules from cyclosporin A-treated cells.


Subject(s)
Cyclosporine/pharmacology , Exosomes/metabolism , Prion Proteins/metabolism , Protein Folding , Secretory Vesicles/metabolism , Animals , CHO Cells , Cricetulus , Humans
16.
BMC Biol ; 15(1): 29, 2017 04 06.
Article in English | MEDLINE | ID: mdl-28385158

ABSTRACT

BACKGROUND: Animals exhibit astonishingly complex behaviors. Studying the subtle features of these behaviors requires quantitative, high-throughput, and accurate systems that can cope with the often rich perplexing data. RESULTS: Here, we present a Multi-Animal Tracker (MAT) that provides a user-friendly, end-to-end solution for imaging, tracking, and analyzing complex behaviors of multiple animals simultaneously. At the core of the tracker is a machine learning algorithm that provides immense flexibility to image various animals (e.g., worms, flies, zebrafish, etc.) under different experimental setups and conditions. Focusing on C. elegans worms, we demonstrate the vast advantages of using this MAT in studying complex behaviors. Beginning with chemotaxis, we show that approximately 100 animals can be tracked simultaneously, providing rich behavioral data. Interestingly, we reveal that worms' directional changes are biased, rather than random - a strategy that significantly enhances chemotaxis performance. Next, we show that worms can integrate environmental information and that directional changes mediate the enhanced chemotaxis towards richer environments. Finally, offering high-throughput and accurate tracking, we show that the system is highly suitable for longitudinal studies of aging- and proteotoxicity-associated locomotion deficits, enabling large-scale drug and genetic screens. CONCLUSIONS: Together, our tracker provides a powerful and simple system to study complex behaviors in a quantitative, high-throughput, and accurate manner.


Subject(s)
Behavior, Animal , Caenorhabditis elegans/physiology , Ethology/methods , Aging/physiology , Algorithms , Animals , Chemotaxis , Machine Learning , Nerve Degeneration/pathology , Proteins/toxicity , Software , Time Factors , Video Recording
17.
Front Neurosci ; 11: 48, 2017.
Article in English | MEDLINE | ID: mdl-28223916

ABSTRACT

Although, protein aggregation and deposition are unifying features of various neurodegenerative disorders, recent studies indicate that different mechanisms can lead to the development of the same malady. Among these, failure in early protein folding and maturation emerge as key mechanistic events that lead to the manifestation of a myriad of illnesses including Alzheimer's disease and prion disorders. Here we delineate the cascade of maturation steps that nascent polypeptides undergo in the secretory pathway to become functional proteins, and the chaperones that supervise and assist this process, focusing on the subgroup of proline cis/trans isomerases. We also describe the chaperones whose failure was found to be an underlying event that initiates the run-up toward neurodegeneration as well as chaperones whose activity impairs protein homeostasis (proteostasis) and thus, promotes the manifestation of these maladies. Finally, we discuss the roles of aggregate deposition sites in the cellular attempt to maintain proteostasis and point at potential targets for therapeutic interventions.

18.
Article in English | MEDLINE | ID: mdl-27864315

ABSTRACT

Maintaining functional protein homeostasis (proteostasis) is a constant challenge in the face of limited protein-folding capacity, environmental threats, and aging. Cells have developed several quality-control mechanisms that assist nascent polypeptides to fold properly, clear misfolded molecules, respond to the accumulation of protein aggregates, and deposit potentially toxic conformers in designated sites. Proteostasis collapse can lead to the development of diseases known as proteinopathies. Here we delineate the current knowledge on the different layers of protein quality-control mechanisms at the organelle and cellular levels with an emphasis on the prion protein (PrP). We also describe how protein quality control is integrated at the organismal level and discuss future perspectives on utilizing proteostasis maintenance as a strategy to develop novel therapies for the treatment of proteinopathies.


Subject(s)
Quality Control , Humans , Unfolded Protein Response
19.
J Cell Sci ; 129(19): 3635-3647, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27550517

ABSTRACT

Limited detoxification capacity often directs aggregation-prone, potentially hazardous, misfolded proteins to be deposited in designated cytosolic compartments known as 'aggresomes'. The roles of aggresomes as cellular quality control centers, and the cellular origin of the deposits contained within these structures, remain to be characterized. Here, we utilized the observation that the prion protein (PrP, also known as PRNP) accumulates in aggresomes following the inhibition of folding chaperones, members of the cyclophilin family, to address these questions. We found that misfolded PrP molecules must pass through the endoplasmic reticulum (ER) in order to be deposited in aggresomes, that the Golgi plays no role in this process and that cytosolic PrP species are not deposited in pre-existing aggresomes. Prior to their deposition in the aggresome, PrP molecules lose the ER localization signal and have to acquire a GPI anchor. Our discoveries indicate that PrP aggresomes are cytosolic overflow deposition centers for the ER quality control mechanisms and highlight the importance of these structures for the maintenance of protein homeostasis within the ER.


Subject(s)
Cytosol/metabolism , Endoplasmic Reticulum/metabolism , Prion Proteins/metabolism , Protein Aggregates , Animals , CHO Cells , Cricetinae , Cricetulus , Cyclosporine/pharmacology , Cytosol/drug effects , Endoplasmic Reticulum/drug effects , Glycosylation , Glycosylphosphatidylinositols/metabolism , Golgi Apparatus/drug effects , Golgi Apparatus/metabolism , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Protein Aggregates/drug effects , Protein Folding/drug effects
20.
Cell Cycle ; 15(7): 877-8, 2016.
Article in English | MEDLINE | ID: mdl-26939744
SELECTION OF CITATIONS
SEARCH DETAIL