Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
Cell ; 139(6): 1157-69, 2009 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-20005808

RESUMEN

The insulin/insulin growth factor (IGF) signaling (IIS) pathway is a key regulator of aging of worms, flies, mice, and likely humans. Delayed aging by IIS reduction protects the nematode C. elegans from toxicity associated with the aggregation of the Alzheimer's disease-linked human peptide, Abeta. We reduced IGF signaling in Alzheimer's model mice and discovered that these animals are protected from Alzheimer's-like disease symptoms, including reduced behavioral impairment, neuroinflammation, and neuronal loss. This protection is correlated with the hyperaggregation of Abeta leading to tightly packed, ordered plaques, suggesting that one aspect of the protection conferred by reduced IGF signaling is the sequestration of soluble Abeta oligomers into dense aggregates of lower toxicity. These findings indicate that the IGF signaling-regulated mechanism that protects from Abeta toxicity is conserved from worms to mammals and point to the modulation of this signaling pathway as a promising strategy for the development of Alzheimer's disease therapy.


Asunto(s)
Factor I del Crecimiento Similar a la Insulina/metabolismo , Longevidad , Transducción de Señal , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/fisiopatología , Péptidos beta-Amiloides/metabolismo , Animales , Humanos , Masculino , Ratones , Ratones Transgénicos , Presenilina-1/genética , Presenilina-1/metabolismo , Receptor IGF Tipo 1/metabolismo
2.
EMBO Rep ; 19(8)2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29945933

RESUMEN

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.


Asunto(s)
Envejecimiento/metabolismo , Caenorhabditis elegans/metabolismo , Caveolas/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Insulina/metabolismo , Transducción de Señal , Animales , Caenorhabditis elegans/genética , Caenorhabditis elegans/ultraestructura , Proteínas de Caenorhabditis elegans/metabolismo , Caveolas/ultraestructura , Caveolina 1/metabolismo , Caveolina 2/metabolismo , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Respuesta al Choque Térmico , Longevidad , Modelos Biológicos , Proteostasis , Interferencia de ARN , Factores de Transcripción/metabolismo , Rayos Ultravioleta
3.
EMBO J ; 34(22): 2820-39, 2015 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-26438723

RESUMEN

Do different neurodegenerative maladies emanate from the failure of a mutual protein folding mechanism? We have addressed this question by comparing mutational patterns that are linked to the manifestation of distinct neurodegenerative disorders and identified similar neurodegeneration-linked proline substitutions in the prion protein and in presenilin 1 that underlie the development of a prion disorder and of familial Alzheimer's disease (fAD), respectively. These substitutions were found to prevent the endoplasmic reticulum (ER)-resident chaperone, cyclophilin B, from assisting presenilin 1 to fold properly, leading to its aggregation, deposition in the ER, reduction of γ-secretase activity, and impaired mitochondrial distribution and function. Similarly, reduced quantities of the processed, active presenilin 1 were observed in brains of cyclophilin B knockout mice. These discoveries imply that reduced cyclophilin activity contributes to the development of distinct neurodegenerative disorders, propose a novel mechanism for the development of certain fAD cases, and support the emerging theme that this disorder can stem from aberrant presenilin 1 function. This study also points at ER chaperones as targets for the development of counter-neurodegeneration therapies.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Sustitución de Aminoácidos , Encéfalo/metabolismo , Presenilina-1/metabolismo , Agregación Patológica de Proteínas/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Animales , Encéfalo/patología , Línea Celular , Ratones , Ratones Noqueados , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Presenilina-1/genética , Prolina/genética , Prolina/metabolismo , Agregación Patológica de Proteínas/genética , Agregación Patológica de Proteínas/patología , Pliegue de Proteína
4.
FASEB J ; 32(3): 1479-1492, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29127190

RESUMEN

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.


Asunto(s)
Ciclosporina/farmacología , Exosomas/metabolismo , Proteínas Priónicas/metabolismo , Pliegue de Proteína , Vesículas Secretoras/metabolismo , Animales , Células CHO , Cricetulus , Humanos
5.
J Cell Sci ; 129(19): 3635-3647, 2016 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-27550517

RESUMEN

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.


Asunto(s)
Citosol/metabolismo , Retículo Endoplásmico/metabolismo , Proteínas Priónicas/metabolismo , Agregado de Proteínas , Animales , Células CHO , Cricetinae , Cricetulus , Ciclosporina/farmacología , Citosol/efectos de los fármacos , Retículo Endoplásmico/efectos de los fármacos , Glicosilación , Glicosilfosfatidilinositoles/metabolismo , Aparato de Golgi/efectos de los fármacos , Aparato de Golgi/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Biológicos , Agregado de Proteínas/efectos de los fármacos , Pliegue de Proteína/efectos de los fármacos
6.
BMC Biol ; 15(1): 29, 2017 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-28385158

RESUMEN

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.


Asunto(s)
Conducta Animal , Caenorhabditis elegans/fisiología , Etología/métodos , Envejecimiento/fisiología , Algoritmos , Animales , Quimiotaxis , Aprendizaje Automático , Degeneración Nerviosa/patología , Proteínas/toxicidad , Programas Informáticos , Factores de Tiempo , Grabación en Video
7.
FASEB J ; 30(4): 1656-69, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26722006

RESUMEN

The discovery that the alteration of aging by reducing the activity of the insulin/IGF-1 signaling (IIS) cascade protects nematodes and mice from neurodegeneration-linked, toxic protein aggregation (proteotoxicity) raises the prospect that IIS inhibitors bear therapeutic potential to counter neurodegenerative diseases. Recently, we reported that NT219, a highly efficient IGF-1 signaling inhibitor, protects model worms from the aggregation of amyloid ß peptide and polyglutamine peptides that are linked to the manifestation of Alzheimer's and Huntington's diseases, respectively. Here, we employed cultured cell systems to investigate whether NT219 promotes protein homeostasis (proteostasis) in mammalian cells and to explore its underlying mechanisms. We found that NT219 enhances the aggregation of misfolded prion protein and promotes its deposition in quality control compartments known as "aggresomes." NT219 also elevates the levels of certain molecular chaperones but, surprisingly, reduces proteasome activity and impairs autophagy. Our findings show that IGF-1 signaling inhibitors in general and NT219 in particular can promote proteostasis in mammalian cells by hyperaggregating hazardous proteins, thereby bearing the potential to postpone the onset and slow the progression of neurodegenerative illnesses in the elderly.-Moll, L., Ben-Gedalya, T., Reuveni, H., Cohen, E. The inhibition of IGF-1 signaling promotes proteostasis by enhancing protein aggregation and deposition.


Asunto(s)
Homeostasis/fisiología , Factor I del Crecimiento Similar a la Insulina/metabolismo , Proteínas/metabolismo , Transducción de Señal/fisiología , Péptidos beta-Amiloides/antagonistas & inhibidores , Péptidos beta-Amiloides/metabolismo , Animales , Western Blotting , Células CHO , Caenorhabditis elegans/efectos de los fármacos , Caenorhabditis elegans/metabolismo , Línea Celular Tumoral , Células Cultivadas , Cricetinae , Cricetulus , Expresión Génica/efectos de los fármacos , Homeostasis/efectos de los fármacos , Ratones , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Células 3T3 NIH , Enfermedades Neurodegenerativas/metabolismo , Compuestos Orgánicos/farmacología , Células PC12 , Priones/antagonistas & inhibidores , Priones/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Pirogalol/análogos & derivados , Pirogalol/farmacología , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Tioamidas/farmacología
8.
Methods ; 68(3): 458-64, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24794346

RESUMEN

Toxicity arising from protein misfolding and aggregation (proteotoxicity) is tightly mechanistically linked to the emergence of late-onset neurodegenerative disorders such as Alzheimer's and Parkinson's diseases. Why these maladies manifest in late stages of life and what mechanisms protect the young organism from disease are key enigmas. The nematode Caenorhabditis elegans offers key advantages that enable systematic exploration of many cell biological and functional aspects of neurodegeneration-linked proteotoxicity. Here we review the abundantly used nematode-based proteotoxicity models and delineate common techniques for the measurement of protein aggregation and rate of proteotoxicity. We also discuss the advantages offered by the worm for genetic screening, drug development and for the exploration of the links between proteotoxicity and the aging process.


Asunto(s)
Envejecimiento/genética , Caenorhabditis elegans/genética , Degeneración Nerviosa/genética , Envejecimiento/patología , Enfermedad de Alzheimer/genética , Animales , Caenorhabditis elegans/fisiología , Humanos , Modelos Animales , Degeneración Nerviosa/patología , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología , Transducción de Señal/genética
9.
Traffic ; 13(5): 635-42, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22280095

RESUMEN

Maintenance of proteome integrity (proteostasis) is essential for cellular and organismal survival. Various cellular mechanisms work to preserve proteostasis by ensuring correct protein maturation and efficient degradation of misfolded and damaged proteins. Despite this cellular effort, under certain circumstances subsets of aggregation-prone proteins escape the quality control surveillance, accumulate within the cell and form insoluble aggregates that can lead to the development of disorders including late-onset neurodegenerative diseases. Cells respond to the appearance of insoluble aggregates by actively transporting them to designated deposition sites where they often undergo degradation. Although several protein aggregate deposition sites have been described and extensively studied, key questions regarding their biological roles and how they are affected by aging remained unanswered. Here we review the recent advances in the field, describe the different subtypes of these cellular compartments and outline the evidence that these structures change their properties over time. Finally, we propose models to explain the possible mechanistic links between aggregate deposition sites, neurodegenerative disorders and the aging process.


Asunto(s)
Envejecimiento , Encéfalo/patología , Animales , Transporte Biológico , Encéfalo/metabolismo , Caenorhabditis elegans , Drosophila melanogaster/metabolismo , Humanos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Modelos Biológicos , Enfermedades Neurodegenerativas/metabolismo , Neuronas/metabolismo , Péptidos/química , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas/química , Ratas
10.
J Neurosci ; 33(14): 6102-11, 2013 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-23554491

RESUMEN

In the nematode Caenorhabditis elegans, the heat shock response (HSR) is regulated at the organismal level by a network of thermosensory neurons that senses elevated temperatures and activates the HSR in remote tissues. Which neuronal receptors are required for this signaling mechanism and in which neurons they function are largely unanswered questions. Here we used worms that were engineered to exhibit RNA interference hypersensitivity in neurons to screen for neuronal receptors that are required for the activation of the HSR and identified a putative G-protein coupled receptor (GPCR) as a novel key component of this mechanism. This gene, which we termed GPCR thermal receptor 1 (gtr-1), is expressed in chemosensory neurons and has no role in heat sensing but is critically required for the induction of genes that encode heat shock proteins in non-neural tissues upon exposure to heat. Surprisingly, the knock-down of gtr-1 by RNA interference protected worms expressing the Alzheimer's-disease-linked aggregative peptide Aß3-42 from proteotoxicity but had no effect on lifespan. This study provides several novel insights: (1) it shows that chemosensory neurons play important roles in the nematode's HSR-regulating mechanism, (2) it shows that lifespan and heat stress resistance are separable, and (3) it strengthens the emerging notion that the ability to respond to heat comes at the expense of protein homeostasis (proteostasis).


Asunto(s)
Regulación de la Expresión Génica/fisiología , Respuesta al Choque Térmico/fisiología , Calor , Neuronas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Sensación Térmica/fisiología , Péptidos beta-Amiloides/metabolismo , Animales , Animales Modificados Genéticamente , Infecciones Bacterianas/prevención & control , Caenorhabditis elegans , Proteínas de Caenorhabditis elegans/genética , Huevos , Regulación de la Expresión Génica/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Respuesta al Choque Térmico/genética , Músculos/metabolismo , Mutación/genética , Parálisis/genética , Fragmentos de Péptidos/metabolismo , Lectinas de Plantas/genética , Lectinas de Plantas/metabolismo , Interferencia de ARN , ARN Mensajero/genética , Receptores Acoplados a Proteínas G/genética , Conducta Sexual Animal , Transducción de Señal/genética , Transducción de Señal/fisiología , Estrés Fisiológico/genética , Estrés Fisiológico/fisiología , Sensación Térmica/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
11.
Nat Aging ; 4(6): 791-813, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38750187

RESUMEN

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.


Asunto(s)
Células Germinativas , Longevidad , Animales , Longevidad/genética , Masculino , Femenino , Células Germinativas/metabolismo , Caenorhabditis elegans/genética , Caenorhabditis elegans/fisiología , Caracteres Sexuales
12.
bioRxiv ; 2024 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-38187630

RESUMEN

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.

13.
J Cell Sci ; 124(Pt 11): 1891-902, 2011 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-21558416

RESUMEN

Despite the activity of cellular quality-control mechanisms, subsets of mature and newly synthesized polypeptides fail to fold properly and form insoluble aggregates. In some cases, protein aggregation leads to the development of human neurodegenerative maladies, including Alzheimer's and prion diseases. Aggregates of misfolded prion protein (PrP), which appear in cells after exposure to the drug cyclosporin A (CsA), and disease-linked PrP mutants have been found to accumulate in juxtanuclear deposition sites termed 'aggresomes'. Recently, it was shown that cells can contain at least two types of deposition sites for misfolded proteins: a dynamic quality-control compartment, which was termed 'JUNQ', and a site for terminally aggregated proteins called 'IPOD'. Here, we show that CsA-induced PrP aggresomes are dynamic structures that form despite intact proteasome activity, recruit chaperones and dynamically exchange PrP molecules with the cytosol. These findings define the CsA-PrP aggresome as a JUNQ-like dynamic quality-control compartment that mediates the refolding or degradation of misfolded proteins. Together, our data suggest that the formation of PrP aggresomes protects cells from proteotoxic stress.


Asunto(s)
Ciclosporina/farmacología , Cuerpos de Inclusión/metabolismo , Priones/metabolismo , Animales , Células CHO , Cricetinae , Cricetulus , Cristalinas/metabolismo , Proteínas HSP70 de Choque Térmico/metabolismo , Humanos , Complejos Multiproteicos/metabolismo , Enfermedades por Prión/metabolismo , Priones/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Pliegue de Proteína , Ubiquitina/metabolismo , Proteínas Ubiquitinadas/metabolismo
14.
Nat Rev Neurosci ; 9(10): 759-67, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18769445

RESUMEN

Distinct human neurodegenerative diseases share remarkably similar temporal emergence patterns, even though different toxic proteins are involved in their onset. Typically, familial neurodegenerative diseases emerge during the fifth decade of life, whereas sporadic cases do not exhibit symptoms earlier than the seventh decade. Recently, mechanistic links between the aging process and toxic protein aggregation, a common hallmark of neurodegenerative diseases, have been revealed. The insulin/insulin-like growth factor 1 (IGF1) signalling pathway - a lifespan, metabolism and stress-resistance regulator - links neurodegeneration to the aging process. Thus, although a reduction of insulin signalling can result in diabetes, its reduction can also increase longevity and delay the onset of protein-aggregation-mediated toxicity. Here we review this apparent paradox and delineate the therapeutic potential of manipulating the insulin/IGF1 signalling pathway for the treatment of neurodegenerative diseases.


Asunto(s)
Envejecimiento/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Insulina/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Animales , Diabetes Mellitus/metabolismo , Diabetes Mellitus/fisiopatología , Humanos , Cuerpos de Inclusión/metabolismo , Cuerpos de Inclusión/patología , Longevidad/fisiología , Transducción de Señal/fisiología
15.
Front Mol Biosci ; 10: 1290118, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38016061

RESUMEN

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.

16.
Aging Cell ; 22(12): e14013, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37897137

RESUMEN

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.


Asunto(s)
Enfermedad de Alzheimer , Caenorhabditis elegans , Animales , Humanos , Caenorhabditis elegans/genética , Péptidos beta-Amiloides/toxicidad , Péptidos beta-Amiloides/metabolismo , Factores de Transcripción/metabolismo , Enfermedad de Alzheimer/tratamiento farmacológico , Envejecimiento
17.
Cell Rep ; 38(6): 110350, 2022 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-35139369

RESUMEN

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.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Unión al ADN/metabolismo , Neuropéptidos/metabolismo , Proteostasis/fisiología , Transducción de Señal/fisiología , Factores de Transcripción/metabolismo , Animales , Caenorhabditis elegans/metabolismo , Homeostasis/fisiología , Péptidos/metabolismo , Proteoma/metabolismo
18.
Biochemistry ; 50(10): 1607-17, 2011 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-21268584

RESUMEN

The process of amyloid-ß (Aß) fibril formation is genetically and pathologically linked to Alzheimer's disease (AD). Thus, a selective and sensitive method for quantifying Aß fibrils in complex biological samples allows a variety of hypotheses to be tested. Herein, we report the basis for a quantitative in vitro kinetic aggregation assay that detects seeding-competent Aß aggregates in mammalian cell culture media, in Caenorhabditis elegans lysate, and in mouse brain homogenate. Sonicated, proteinase K-treated Aß fibril-containing tissue homogenates or cell culture media were added to an initially monomeric Aß(1-40) reporter peptide to seed an in vitro nucleated aggregation reaction. The reduction in the half-time (t(50)) of the amyloid growth phase is proportional to the quantity of seeding-competent Aß aggregates present in the biological sample. An ion-exchange resin amyloid isolation strategy from complex biological samples is demonstrated as an alternative for improving the sensitivity and linearity of the kinetic aggregation assay.


Asunto(s)
Amiloide/análisis , Caenorhabditis elegans/química , Amiloide/metabolismo , Péptidos beta-Amiloides/análisis , Animales , Química Encefálica , Endopeptidasa K/metabolismo , Cinética , Ratones , Fragmentos de Péptidos/análisis
19.
PLoS One ; 16(7): e0243522, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34197476

RESUMEN

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.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/fisiología , Proteínas de Unión al ADN/metabolismo , Longevidad , Proteostasis/fisiología , Factores de Transcripción/metabolismo , Animales , Caenorhabditis elegans/crecimiento & desarrollo , Proteínas de Caenorhabditis elegans/antagonistas & inhibidores , Proteínas de Caenorhabditis elegans/genética , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , Factores de Transcripción Forkhead/metabolismo , Larva/efectos de los fármacos , Larva/crecimiento & desarrollo , Larva/metabolismo , Péptidos/farmacología , Interferencia de ARN , ARN Bicatenario/metabolismo , Ribonucleasa III/antagonistas & inhibidores , Ribonucleasa III/genética , Ribonucleasa III/metabolismo , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/genética
20.
Genetics ; 215(4): 889-901, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32759342

RESUMEN

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.


Asunto(s)
Caenorhabditis elegans/fisiología , Proteínas HSP70 de Choque Térmico/metabolismo , Homeostasis , Proteoma/metabolismo , Deficiencias en la Proteostasis/fisiopatología , Proteostasis , Estrés Fisiológico , Animales , Humanos , Pliegue de Proteína , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA