Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell Mol Life Sci ; 80(11): 342, 2023 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-37904059

RESUMEN

Arsenic and antimony are metalloids with profound effects on biological systems and human health. Both elements are toxic to cells and organisms, and exposure is associated with several pathological conditions including cancer and neurodegenerative disorders. At the same time, arsenic- and antimony-containing compounds are used in the treatment of multiple diseases. Although these metalloids can both cause and cure disease, their modes of molecular action are incompletely understood. The past decades have seen major advances in our understanding of arsenic and antimony toxicity, emphasizing genotoxicity and proteotoxicity as key contributors to pathogenesis. In this review, we highlight mechanisms by which arsenic and antimony cause toxicity, focusing on their genotoxic and proteotoxic effects. The mechanisms used by cells to maintain proteostasis during metalloid exposure are also described. Furthermore, we address how metalloid-induced proteotoxicity may promote neurodegenerative disease and how genotoxicity and proteotoxicity may be interrelated and together contribute to proteinopathies. A deeper understanding of cellular toxicity and response mechanisms and their links to pathogenesis may promote the development of strategies for both disease prevention and treatment.


Asunto(s)
Arsénico , Metaloides , Enfermedades Neurodegenerativas , Humanos , Arsénico/toxicidad , Antimonio/toxicidad , Enfermedades Neurodegenerativas/inducido químicamente , Daño del ADN
2.
Proc Natl Acad Sci U S A ; 118(30)2021 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-34290138

RESUMEN

Nuclear envelope budding (NEB) is a recently discovered alternative pathway for nucleocytoplasmic communication distinct from the movement of material through the nuclear pore complex. Through quantitative electron microscopy and tomography, we demonstrate how NEB is evolutionarily conserved from early protists to human cells. In the yeast Saccharomyces cerevisiae, NEB events occur with higher frequency during heat shock, upon exposure to arsenite or hydrogen peroxide, and when the proteasome is inhibited. Yeast cells treated with azetidine-2-carboxylic acid, a proline analog that induces protein misfolding, display the most dramatic increase in NEB, suggesting a causal link to protein quality control. This link was further supported by both localization of ubiquitin and Hsp104 to protein aggregates and NEB events, and the evolution of these structures during heat shock. We hypothesize that NEB is part of normal cellular physiology in a vast range of species and that in S. cerevisiae NEB comprises a stress response aiding the transport of protein aggregates across the nuclear envelope.


Asunto(s)
Ácido Azetidinocarboxílico/toxicidad , Respuesta al Choque Térmico , Membrana Nuclear/fisiología , Pliegue de Proteína , Proteostasis/efectos de los fármacos , Proteínas de Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/crecimiento & desarrollo , Arsenitos/toxicidad , Peróxido de Hidrógeno/toxicidad , Membrana Nuclear/efectos de los fármacos , Saccharomyces cerevisiae/efectos de los fármacos , Proteínas de Saccharomyces cerevisiae/efectos de los fármacos , Proteínas de Saccharomyces cerevisiae/metabolismo , Compuestos de Sodio/toxicidad , Ubiquitina/metabolismo , Ubiquitinación
3.
J Biol Chem ; 298(12): 102680, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36356902

RESUMEN

The poisonous metalloid arsenite induces widespread misfolding and aggregation of nascent proteins in vivo, and this mode of toxic action might underlie its suspected role in the pathology of certain protein misfolding diseases. Evolutionarily conserved protein quality-control systems protect cells against arsenite-mediated proteotoxicity, and herein, we systematically assessed the contribution of the ubiquitin-proteasome system, the autophagy-vacuole pathway, and chaperone-mediated disaggregation to the clearance of arsenite-induced protein aggregates in Saccharomyces cerevisiae. We show that the ubiquitin-proteasome system is the main pathway that clears aggregates formed during arsenite stress and that cells depend on this pathway for optimal growth. The autophagy-vacuole pathway and chaperone-mediated disaggregation both contribute to clearance, but their roles appear less prominent than the ubiquitin-proteasome system. Our in vitro assays with purified components of the yeast disaggregating machinery demonstrated that chaperone binding to aggregates formed in the presence of arsenite is impaired. Hsp104 and Hsp70 chaperone activity was unaffected by arsenite, suggesting that this metalloid influences aggregate structure, making them less accessible for chaperone-mediated disaggregation. We further show that the defect in chaperone-mediated refolding of a model protein was abrogated in a cysteine-free version of the substrate, suggesting that arsenite directly modifies cysteines in non-native target proteins. In conclusion, our study sheds novel light on the differential contributions of protein quality-control systems to aggregate clearance and cell proliferation and extends our understanding of how these systems operate during arsenite stress.


Asunto(s)
Arsenitos , Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Agregado de Proteínas , Arsenitos/farmacología , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Chaperonas Moleculares/metabolismo , Proteínas HSP70 de Choque Térmico/metabolismo , Ubiquitina/metabolismo , Autofagia , Proteínas de Choque Térmico/metabolismo
4.
J Cell Sci ; 134(11)2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-34085697

RESUMEN

The toxic metalloid arsenic causes widespread misfolding and aggregation of cellular proteins. How these protein aggregates are formed in vivo, the mechanisms by which they affect cells and how cells prevent their accumulation is not fully understood. To find components involved in these processes, we performed a genome-wide imaging screen and identified Saccharomyces cerevisiae deletion mutants with either enhanced or reduced protein aggregation levels during arsenite exposure. We show that many of the identified factors are crucial to safeguard protein homeostasis (proteostasis) and to protect cells against arsenite toxicity. The hits were enriched for various functions including protein biosynthesis and transcription, and dedicated follow-up experiments highlight the importance of accurate transcriptional and translational control for mitigating protein aggregation and toxicity during arsenite stress. Some of the hits are associated with pathological conditions, suggesting that arsenite-induced protein aggregation may affect disease processes. The broad network of cellular systems that impinge on proteostasis during arsenic stress identified in this current study provides a valuable resource and a framework for further elucidation of the mechanistic details of metalloid toxicity and pathogenesis. This article has an associated First Person interview with the first authors of the paper.


Asunto(s)
Arsénico , Arsenitos , Proteínas de Saccharomyces cerevisiae , Arsenitos/toxicidad , Agregado de Proteínas , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética
5.
Nucleic Acids Res ; 49(16): 9280-9293, 2021 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-34387667

RESUMEN

Activator proteins 1 (AP-1) comprise one of the largest families of eukaryotic basic leucine zipper transcription factors. Despite advances in the characterization of AP-1 DNA-binding sites, our ability to predict new binding sites and explain how the proteins achieve different gene expression levels remains limited. Here we address the role of sequence-specific DNA flexibility for stability and specific binding of AP-1 factors, using microsecond-long molecular dynamics simulations. As a model system, we employ yeast AP-1 factor Yap1 binding to three different response elements from two genetic environments. Our data show that Yap1 actively exploits the sequence-specific flexibility of DNA within the response element to form stable protein-DNA complexes. The stability also depends on the four to six flanking nucleotides, adjacent to the response elements. The flanking sequences modulate the conformational adaptability of the response element, making it more shape-efficient to form specific contacts with the protein. Bioinformatics analysis of differential expression of the studied genes supports our conclusions: the stability of Yap1-DNA complexes, modulated by the flanking environment, influences the gene expression levels. Our results provide new insights into mechanisms of protein-DNA recognition and the biological regulation of gene expression levels in eukaryotes.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , ADN/genética , Proteínas de Saccharomyces cerevisiae/genética , Factor de Transcripción AP-1/genética , Factores de Transcripción/genética , Secuencia de Bases/genética , Sitios de Unión/genética , ADN/ultraestructura , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/ultraestructura , Regulación de la Expresión Génica/genética , Sustancias Macromoleculares/ultraestructura , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/ultraestructura , Simulación de Dinámica Molecular , Elementos de Respuesta/genética , Proteínas de Saccharomyces cerevisiae/ultraestructura , Factor de Transcripción AP-1/ultraestructura , Factores de Transcripción/ultraestructura , Proteínas Señalizadoras YAP
6.
FEMS Yeast Res ; 22(1)2022 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-35323907

RESUMEN

In a high-throughput yeast two-hybrid screen of predicted coiled-coil motif interactions in the Saccharomyces cerevisiae proteome, the protein Etp1 was found to interact with the yeast AP-1-like transcription factors Yap8, Yap1 and Yap6. Yap8 plays a crucial role during arsenic stress since it regulates expression of the resistance genes ACR2 and ACR3. The function of Etp1 is not well understood but the protein has been implicated in transcription and protein turnover during ethanol stress, and the etp1∆ mutant is sensitive to ethanol. In this current study, we investigated whether Etp1 is implicated in Yap8-dependent functions. We show that Etp1 is required for optimal growth in the presence of trivalent arsenite and for optimal expression of the arsenite export protein encoded by ACR3. Since Yap8 is the only known transcription factor that regulates ACR3 expression, we investigated whether Etp1 regulates Yap8. Yap8 ubiquitination, stability, nuclear localization and ACR3 promoter association were unaffected in etp1∆ cells, indicating that Etp1 affects ACR3 expression independently of Yap8. Thus, Etp1 impacts gene expression under arsenic and other stress conditions but the mechanistic details remain to be elucidated.


Asunto(s)
Arsénico , Arsenitos , Proteínas de Saccharomyces cerevisiae , Arsénico/metabolismo , Arsenitos/metabolismo , Arsenitos/farmacología , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Etanol/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
7.
Nucleic Acids Res ; 48(10): 5426-5441, 2020 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-32356892

RESUMEN

Activator protein 1 (AP-1) is one of the largest families of basic leucine zipper (bZIP) transcription factors in eukaryotic cells. How AP-1 proteins achieve target DNA binding specificity remains elusive. In Saccharomyces cerevisiae, the AP-1-like protein (Yap) family comprises eight members (Yap1 to Yap8) that display distinct genomic target sites despite high sequence homology of their DNA binding bZIP domains. In contrast to the other members of the Yap family, which preferentially bind to short (7-8 bp) DNA motifs, Yap8 binds to an unusually long DNA motif (13 bp). It has been unclear what determines this unique specificity of Yap8. In this work, we use molecular and biochemical analyses combined with computer-based structural design and molecular dynamics simulations of Yap8-DNA interactions to better understand the structural basis of DNA binding specificity determinants. We identify specific residues in the N-terminal tail preceding the basic region, which define stable association of Yap8 with its target promoter. We propose that the N-terminal tail directly interacts with DNA and stabilizes Yap8 binding to the 13 bp motif. Thus, beside the core basic region, the adjacent N-terminal region contributes to alternative DNA binding selectivity within the AP-1 family.


Asunto(s)
Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/química , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , ADN de Hongos/metabolismo , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/metabolismo , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , ADN de Hongos/química , Proteínas de Transporte de Membrana/genética , Simulación de Dinámica Molecular , Mutación , Regiones Promotoras Genéticas , Unión Proteica , Elementos de Respuesta , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética
8.
FEMS Yeast Res ; 21(1)2021 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-33355361

RESUMEN

The Saccharomyces cerevisiae MBOAT O-acyltransferase Gup1 is involved in many processes, including cell wall and membrane composition and integrity, and acetic acid-induced cell death. Gup1 was previously shown to interact physically with the mitochondrial membrane VDAC (Voltage-Dependent Anion Channel) protein Por1 and the ammonium transceptor Mep2. By co-immunoprecipitation, the eisosome core component Pil1 was identified as a novel physical interaction partner of Gup1. The expression of PIL1 and Pil1 protein levels were found to be unaffected by GUP1 deletion. In ∆gup1 cells, Pil1 was distributed in dots (likely representing eisosomes) in the membrane, identically to wt cells. However, ∆gup1 cells presented 50% less Pil1-GFP dots/eisosomes, suggesting that Gup1 is important for eisosome formation. The two proteins also interact genetically in the maintenance of cell wall integrity, and during arsenite and acetic acid exposure. We show that Δgup1 Δpil1 cells take up more arsenite than wt and are extremely sensitive to arsenite and to acetic acid treatments. The latter causes a severe apoptotic wt-like cell death phenotype, epistatically reverting the ∆gup1 necrotic type of death. Gup1 and Pil1 are thus physically, genetically and functionally connected.


Asunto(s)
Proteínas de Transporte de Membrana/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Apoptosis , Membrana Celular/metabolismo , Pared Celular/metabolismo , Proteínas de Transporte de Membrana/genética , Saccharomyces cerevisiae/enzimología
9.
Int J Mol Sci ; 22(21)2021 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-34768886

RESUMEN

Exposure to heavy metals, including arsenic and cadmium, is associated with neurodegenerative disorders such as Parkinson's disease. However, the mechanistic details of how these metals contribute to pathogenesis are not well understood. To search for underlying mechanisms involving α-synuclein, the protein that forms amyloids in Parkinson's disease, we here assessed the effects of arsenic and cadmium on α-synuclein amyloid formation in vitro and in Saccharomyces cerevisiae (budding yeast) cells. Atomic force microscopy experiments with acetylated human α-synuclein demonstrated that amyloid fibers formed in the presence of the metals have a different fiber pitch compared to those formed without metals. Both metal ions become incorporated into the amyloid fibers, and cadmium also accelerated the nucleation step in the amyloid formation process, likely via binding to intermediate species. Fluorescence microscopy analyses of yeast cells expressing fluorescently tagged α-synuclein demonstrated that arsenic and cadmium affected the distribution of α-synuclein aggregates within the cells, reduced aggregate clearance, and aggravated α-synuclein toxicity. Taken together, our in vitro data demonstrate that interactions between these two metals and α-synuclein modulate the resulting amyloid fiber structures, which, in turn, might relate to the observed effects in the yeast cells. Whilst our study advances our understanding of how these metals affect α-synuclein biophysics, further in vitro characterization as well as human cell studies are desired to fully appreciate their role in the progression of Parkinson's disease.


Asunto(s)
Metales Pesados/toxicidad , Enfermedades Neurodegenerativas , alfa-Sinucleína/metabolismo , Amiloide/metabolismo , Arsenitos/toxicidad , Cadmio/toxicidad , Línea Celular/metabolismo , Enfermedades Neurodegenerativas/etiología , Enfermedades Neurodegenerativas/metabolismo , Enfermedad de Parkinson/etiología , Enfermedad de Parkinson/metabolismo , Saccharomyces cerevisiae/metabolismo , alfa-Sinucleína/efectos de los fármacos
10.
Curr Genet ; 64(1): 177-181, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28936749

RESUMEN

Cadmium is a highly poisonous metal and a human carcinogen, but the molecular mechanisms underlying its cellular toxicity are not fully understood. Recent findings in yeast cells indicate that cadmium exerts its deleterious effects by inducing widespread misfolding and aggregation of nascent proteins. Here, we discuss this novel mode of toxic heavy metal action and propose a mechanism by which molecular chaperones may reduce the damaging effects of heavy metal ions on protein structures.


Asunto(s)
Agregado de Proteínas , Agregación Patológica de Proteínas , Pliegue de Proteína , Proteínas/química , Proteínas/metabolismo , Animales , Cadmio/metabolismo , Cadmio/toxicidad , Intoxicación por Metales Pesados , Humanos , Chaperonas Moleculares/química , Chaperonas Moleculares/metabolismo , Proteínas Priónicas/química , Proteínas Priónicas/metabolismo , Unión Proteica , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/metabolismo
11.
Mol Syst Biol ; 12(12): 892, 2016 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-27979908

RESUMEN

A major rationale for the advocacy of epigenetically mediated adaptive responses is that they facilitate faster adaptation to environmental challenges. This motivated us to develop a theoretical-experimental framework for disclosing the presence of such adaptation-speeding mechanisms in an experimental evolution setting circumventing the need for pursuing costly mutation-accumulation experiments. To this end, we exposed clonal populations of budding yeast to a whole range of stressors. By growth phenotyping, we found that almost complete adaptation to arsenic emerged after a few mitotic cell divisions without involving any phenotypic plasticity. Causative mutations were identified by deep sequencing of the arsenic-adapted populations and reconstructed for validation. Mutation effects on growth phenotypes, and the associated mutational target sizes were quantified and embedded in data-driven individual-based evolutionary population models. We found that the experimentally observed homogeneity of adaptation speed and heterogeneity of molecular solutions could only be accounted for if the mutation rate had been near estimates of the basal mutation rate. The ultrafast adaptation could be fully explained by extensive positive pleiotropy such that all beneficial mutations dramatically enhanced multiple fitness components in concert. As our approach can be exploited across a range of model organisms exposed to a variety of environmental challenges, it may be used for determining the importance of epigenetic adaptation-speeding mechanisms in general.


Asunto(s)
Arsénico/farmacología , Proteínas Bacterianas/genética , Epigénesis Genética , Mutación , Saccharomycetales/crecimiento & desarrollo , Adaptación Fisiológica , Evolución Molecular , Aptitud Genética , Secuenciación de Nucleótidos de Alto Rendimiento , Modelos Genéticos , Saccharomycetales/efectos de los fármacos , Saccharomycetales/genética , Selección Genética , Análisis de Secuencia de ADN , Biología de Sistemas/métodos
12.
Biochim Biophys Acta ; 1840(5): 1482-91, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24076236

RESUMEN

BACKGROUND: The yeast Saccharomyces cerevisiae provides unique opportunities to study roles and regulation of aqua/glyceroporins using frontline tools of genetics and genomics as well as molecular cell and systems biology. SCOPE OF REVIEW: S. cerevisiae has two similar orthodox aquaporins. Based on phenotypes mediated by gene deletion or overexpression as well as on their expression pattern, the yeast aquaporins play important roles in key aspects of yeast biology: establishment of freeze tolerance, during spore formation as well as determination of cell surface properties for substrate adhesion and colony formation. Exactly how the aquaporins perform those roles and the mechanisms that regulate their function under such conditions remain to be elucidated. S. cerevisiae also has two different aquaglyceroporins. While the role of one of them, Yfl054c, remains to be determined, Fps1 plays critical roles in osmoregulation by controlling the accumulation of the osmolyte glycerol. Fps1 communicates with two osmo-sensing MAPK signalling pathways to perform its functions but the details of Fps1 regulation remain to be determined. MAJOR CONCLUSIONS: Several phenotypes associated with aqua/glyceroporin function in yeasts have been established. However, how water and glycerol transport contribute to the observed effects is not understood in detail. Also many of the basic principles of regulation of yeast aqua/glyceroporins remain to be elucidated. GENERAL SIGNIFICANCE: Studying the yeast aquaporins and aquaglyceroporins offers rich insight into the life style, evolution and adaptive responses of yeast and rewards us with discoveries of unexpected roles and regulatory mechanisms of members of this ancient protein family. This article is part of a Special Issue entitled Aquaporins.


Asunto(s)
Acuagliceroporinas/fisiología , Acuaporinas/fisiología , Proteínas de Saccharomyces cerevisiae/fisiología , Adaptación Fisiológica/fisiología , Secuencia de Aminoácidos , Acuagliceroporinas/química , Acuaporinas/química , Congelación , Datos de Secuencia Molecular
13.
Biochim Biophys Acta ; 1839(11): 1295-306, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25234620

RESUMEN

All organisms need to sense and respond to a range of stress conditions. In this study, we used transcriptional profiling to identify genes and cellular processes that are responsive during arsenite and tert-butyl hydroperoxide exposure in Kluyveromyces lactis. Many arsenite-responsive genes encode proteins involved in redox processes, protein folding and stabilization, and transmembrane transport. The majority of peroxide-responsive genes encode functions related to transcription, translation, redox processes, metabolism and transport. A substantial number of these stress-regulated genes contain binding motifs for the AP-1 like transcription factors KlYap1 and KlYap8. We demonstrate that KlYap8 binds to and regulates gene expression through a 13 base-pair promoter motif, and that KlYap8 provides protection against arsenite, antimonite, cadmium and peroxide toxicity. Direct transport assays show that Klyap8Δ cells accumulate more arsenic and cadmium than wild type cells and that the Klyap8Δ mutant is defective in arsenic and cadmium export. KlYap8 regulates gene expression in response to both arsenite and peroxide, and might cooperate with KlYap1 in regulation of specific gene targets. Comparison of KlYap8 with its Saccharomyces cerevisiae orthologue ScYap8 indicates that KlYap8 senses and responds to multiple stress signals whereas ScYap8 is only involved in the response to arsenite and antimonite. Thus, our data suggest that functional specialization of ScYap8 has occurred after the whole genome duplication event. This is the first genome-wide stress response analysis in K. lactis and the first demonstration of KlYap8 function.


Asunto(s)
Arsenitos/farmacología , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/fisiología , Proteínas Fúngicas/fisiología , Regulación Fúngica de la Expresión Génica/efectos de los fármacos , Peróxido de Hidrógeno/farmacología , Kluyveromyces/efectos de los fármacos , Kluyveromyces/genética , Estrés Fisiológico/genética , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Proteínas Fúngicas/genética , Kluyveromyces/metabolismo , Análisis por Micromatrices , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/genética , Transcriptoma
14.
Mol Microbiol ; 92(6): 1343-56, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24798644

RESUMEN

Arsenic has a dual role as causative and curative agent of human disease. Therefore, there is considerable interest in elucidating arsenic toxicity and detoxification mechanisms. By an ensemble modelling approach, we identified a best parsimonious mathematical model which recapitulates and predicts intracellular arsenic dynamics for different conditions and mutants, thereby providing novel insights into arsenic toxicity and detoxification mechanisms in yeast, which could partly be confirmed experimentally by dedicated experiments. Specifically, our analyses suggest that: (i) arsenic is mainly protein-bound during short-term (acute) exposure, whereas glutathione-conjugated arsenic dominates during long-term (chronic) exposure, (ii) arsenic is not stably retained, but can leave the vacuole via an export mechanism, and (iii) Fps1 is controlled by Hog1-dependent and Hog1-independent mechanisms during arsenite stress. Our results challenge glutathione depletion as a key mechanism for arsenic toxicity and instead suggest that (iv) increased glutathione biosynthesis protects the proteome against the damaging effects of arsenic and that (v) widespread protein inactivation contributes to the toxicity of this metalloid. Our work in yeast may prove useful to elucidate similar mechanisms in higher eukaryotes and have implications for the use of arsenic in medical therapy.


Asunto(s)
Arsénico/metabolismo , Modelos Teóricos , Saccharomyces cerevisiae/metabolismo , Biotransformación , Inactivación Metabólica
15.
Fungal Genet Biol ; 74: 45-58, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25483129

RESUMEN

Although suggested, the involvement of the HOG pathway in adaptation processes in extremely halotolerant fungus Hortaea werneckii has never been specifically demonstrated. Here, we show that the H. werneckii HOG pathway is very robust, and that it includes two functionally redundant MAPK homologues, HwHog1A and HwHog1B, that show osmolyte-type-dependent phosphorylation. Inhibition of HwHog1 kinase activity with the ATP analogue BPTIP restricts H. werneckii colony growth at 3.0M NaCl, KCl and sorbitol, most likely due to restricted cell division. On the other hand, HwHog1-regulated transcription of a selected group of genes (HwSTL1, HwGUT2, HwOPI3, HwGDH1, HwUGP1, HwGPD1) is an osmolyte-specific process that is important for induction of gene transcription with high NaCl, for regulation of specific genes with high sorbitol, and has no role in KCl stressed cells. Survival of H. werneckii at moderate NaCl and KCl concentrations is not dependent on HwHog1 activity or the calcineurin pathway, and thus alternative mechanisms must exist. The HOG pathway described here is vital for the extreme osmotolerance of H. werneckii, and its regulation shows important differences from the homologue pathways characterised in other mesophilic and halotolerant fungi.


Asunto(s)
Adaptación Fisiológica , Ascomicetos/enzimología , Proteínas Fúngicas/fisiología , Regulación Fúngica de la Expresión Génica , Proteínas Quinasas Activadas por Mitógenos/fisiología , Adaptación Fisiológica/genética , Ascomicetos/efectos de los fármacos , Ascomicetos/genética , Secuencia de Bases , Calcineurina/metabolismo , Simulación por Computador , Proteínas Fúngicas/genética , Redes y Vías Metabólicas/genética , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/genética , Datos de Secuencia Molecular , Concentración Osmolar , Cloruro de Potasio/farmacología , Cloruro de Sodio/farmacología , Sorbitol/farmacología
16.
J Cell Sci ; 125(Pt 21): 5073-83, 2012 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-22946053

RESUMEN

Several metals and metalloids profoundly affect biological systems, but their impact on the proteome and mechanisms of toxicity are not fully understood. Here, we demonstrate that arsenite causes protein aggregation in Saccharomyces cerevisiae. Various molecular chaperones were found to be associated with arsenite-induced aggregates indicating that this metalloid promotes protein misfolding. Using in vivo and in vitro assays, we show that proteins in the process of synthesis/folding are particularly sensitive to arsenite-induced aggregation, that arsenite interferes with protein folding by acting on unfolded polypeptides, and that arsenite directly inhibits chaperone activity. Thus, folding inhibition contributes to arsenite toxicity in two ways: by aggregate formation and by chaperone inhibition. Importantly, arsenite-induced protein aggregates can act as seeds committing other, labile proteins to misfold and aggregate. Our findings describe a novel mechanism of toxicity that may explain the suggested role of this metalloid in the etiology and pathogenesis of protein folding disorders associated with arsenic poisoning.


Asunto(s)
Arsenitos/farmacología , Proteínas de Choque Térmico/metabolismo , Pliegue de Proteína/efectos de los fármacos , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/efectos de los fármacos , Gránulos Citoplasmáticos/metabolismo , Proteínas de Choque Térmico/antagonistas & inhibidores , Luciferasas de Luciérnaga/biosíntesis , Chaperonas Moleculares/antagonistas & inhibidores , Chaperonas Moleculares/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Biosíntesis de Proteínas/efectos de los fármacos , Proteínas Recombinantes/biosíntesis , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/antagonistas & inhibidores
17.
Curr Genet ; 60(3): 193-200, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24643376

RESUMEN

Chemical molecules that inhibit protein kinase activity are important tools to assess the functions of protein kinases in living cells. To develop, test and characterize novel inhibitors, a convenient and reproducible kinase assay is of importance. Here, we applied a biotinylated peptide-based method to assess adenosine triphosphate-competitive inhibitors that target the yeast kinases Hog1, Elm1 and Elm1-as. The peptide substrates contained 13 amino acids, encompassing the consensus sequence surrounding the phosphorylation site. To test whether the lack of distal sites affects inhibitor efficacy, we compared the peptide-based assay with an assay using full-length protein as substrate. Similar inhibitor efficiencies were obtained irrespective of whether peptide or full-length protein was used as kinase substrates. Thus, we demonstrate that the peptide substrates used previously (Dinér et al. in PLoS One 6(5):e20012, 2011) give accurate results compared with protein substrates. We also show that the peptide-based method is suitable for selectivity assays and for inhibitor screening. The use of biotinylated peptide substrates provides a simple and reliable assay for protein kinase inhibitor characterization. The utility of this approach is discussed.


Asunto(s)
Péptidos/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Quinasas/metabolismo , Levaduras/efectos de los fármacos , Levaduras/metabolismo , Relación Dosis-Respuesta a Droga , Concentración 50 Inhibidora , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Inhibidores de Proteínas Quinasas/química , Saccharomyces cerevisiae/efectos de los fármacos , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/antagonistas & inhibidores , Proteínas de Saccharomyces cerevisiae/metabolismo , Bibliotecas de Moléculas Pequeñas , Levaduras/genética
18.
J Biol Chem ; 287(28): 23562-70, 2012 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-22593571

RESUMEN

Aquaglyceroporins are transmembrane proteins belonging to the family of aquaporins, which facilitate the passage of specific uncharged solutes across membranes of cells. The yeast aquaglyceroporin Fps1 is important for osmoadaptation by regulating intracellular glycerol levels during changes in external osmolarity. Upon high osmolarity conditions, yeast accumulates glycerol by increased production of the osmolyte and by restricting glycerol efflux through Fps1. The extended cytosolic termini of Fps1 contain short domains that are important for regulating glycerol flux through the channel. Here we show that the transmembrane core of the protein plays an equally important role. The evidence is based on results from an intragenic suppressor mutation screen and domain swapping between the regulated variant of Fps1 from Saccharomyces cerevisiae and the hyperactive Fps1 ortholog from Ashbya gossypii. This suggests a novel mechanism for regulation of glycerol flux in yeast, where the termini alone are not sufficient to restrict Fps1 transport. We propose that glycerol flux through the channel is regulated by interplay between the transmembrane helices and the termini. This mechanism enables yeast cells to fine-tune intracellular glycerol levels at a wide range of extracellular osmolarities.


Asunto(s)
Acuagliceroporinas/metabolismo , Eremothecium/metabolismo , Glicerol/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Secuencia de Aminoácidos , Acuagliceroporinas/química , Acuagliceroporinas/genética , Sitios de Unión/genética , Transporte Biológico , Eremothecium/genética , Prueba de Complementación Genética , Glicina/genética , Glicina/metabolismo , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Viabilidad Microbiana/efectos de los fármacos , Viabilidad Microbiana/genética , Modelos Moleculares , Datos de Secuencia Molecular , Mutación , Concentración Osmolar , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/crecimiento & desarrollo , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética , Homología de Secuencia de Aminoácido , Cloruro de Sodio/farmacología , Equilibrio Hidroelectrolítico/genética
19.
Mol Microbiol ; 84(6): 1177-88, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22554109

RESUMEN

Arsenic is an environmental toxin and a worldwide health hazard. Since this metalloid is ubiquitous in nature, virtually all living organisms require systems for detoxification and tolerance acquisition. Here, we show that during chronic exposure to arsenite [As(III)], Saccharomyces cerevisiae (budding yeast) exports and accumulates the low-molecular-weight thiol molecule glutathione (GSH) outside of cells. Extracellular accumulation of the arsenite triglutathione complex As(GS)3 was also detected and direct transport assays demonstrate that As(GS)3 does not readily enter cells. Yeast cells with increased extracellular GSH levels accumulate less arsenic and display improved growth when challenged with As(III). Conversely, cells defective in export and extracellular accumulation of GSH are As(III) sensitive. Taken together, our data are consistent with a novel detoxification mechanism in which GSH is exported to protect yeast cells from arsenite toxicity by preventing its uptake.


Asunto(s)
Antifúngicos/antagonistas & inhibidores , Antifúngicos/metabolismo , Arsenitos/antagonistas & inhibidores , Arsenitos/metabolismo , Glutatión/metabolismo , Saccharomyces cerevisiae/efectos de los fármacos , Saccharomyces cerevisiae/metabolismo , Inactivación Metabólica , Saccharomyces cerevisiae/crecimiento & desarrollo
20.
Mol Microbiol ; 85(6): 1204-18, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22779703

RESUMEN

Leishmania major aquaglyceroporin (LmjAQP1) adventitiously facilitates the uptake of antimonite [Sb(III)], an active form of Pentostam® or Glucantime®, which are the first line of defence against all forms of leishmaniasis. The present paper shows that LmjAQP1 activity is modulated by the mitogen-activated protein kinase, LmjMPK2. Leishmania parasites coexpressing LmjAQP1 and LmjMPK2 show increased Sb(III) uptake and increased Sb(III) sensitivity. When subjected to a hypo-osmotic stress, these cells show faster volume recovery than cells expressing LmjAQP1 alone. LmjAQP1 is phosphorylated in vivo at Thr-197 and this phosphorylation requires LmjMPK2 activity. Lys-42 of LmjMPK2 is critical for its kinase activity. Cells expressing altered T197A LmjAQP1 or K42A LmjMPK2 showed decreased Sb(III) influx and a slower volume recovery than cells expressing wild-type proteins. Phosphorylation of LmjAQP1 led to a decrease in its turnover rate affecting LmjAQP1 activity. Although LmjAQP1 is localized to the flagellum of promastigotes, upon phosphorylation, it is relocalized to the entire surface of the parasite. Leishmania mexicana promastigotes with an MPK2 deletion showed reduced Sb(III) uptake and slower volume recovery than wild-type cells. This is the first report where a parasite aquaglyceroporin activity is post-translationally modulated by a mitogen-activated protein kinase.


Asunto(s)
Acuaporina 1/metabolismo , Leishmania major/enzimología , Leishmania major/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Antimonio/metabolismo , Antiprotozoarios/metabolismo , Eliminación de Gen , Leishmania major/efectos de los fármacos , Leishmania mexicana/enzimología , Leishmania mexicana/genética , Pruebas de Sensibilidad Parasitaria
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA