Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Curr Issues Mol Biol ; 36: 63-66, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31559970

RESUMEN

Prion diseases or transmissible spongiform encephalopathies (TSEs) are fatal neurological diseases that include Creutzfeldt-Jakob disease (CJD) in humans, scrapie in sheep and goats, bovine spongiform encephalopathy (BSE) in cattle, camel spongiform encephalopathy (CSE) in camels and chronic wasting disease (CWD) in cervids. A key event in prion diseases is the conversion of the cellular, host-encoded prion protein (PrPC) to its abnormal isoform (PrPSc) predominantly in the central nervous system of the infected host (Aguzzi et al., 2004). These diseases are transmissible under some circumstances, but unlike other transmissible disorders, prion diseases can also be caused by mutations in the host gene. The mechanism of prion spread among sheep and goats that develop natural scrapie is unknown. CWD, transmissible mink encephalopathy (TME), BSE, feline spongiform encephalopathy (FSE), and exotic ungulate encephalopathy (EUE) are all thought to occur after the consumption of prion-infected material. Most cases of human prion disease occur from unknown reasons, and greater than 20 mutations in the prion protein (PrP) gene may lead to inherited prion disease. In other instances, prion diseases are contracted by exposure to prion infectivity. These considerations raise the question of how a mere protein aggregate can bypass mucosal barriers, circumvent innate and adoptive immunity, and traverse the blood-brain barrier to give rise to brain disease. Here, we will briefly introduce a few topics in current prion studies.


Asunto(s)
Enfermedades por Prión/genética , Priones/metabolismo , Deficiencias en la Proteostasis/metabolismo , Enfermedad de Alzheimer/metabolismo , Animales , Inocuidad de los Alimentos , Humanos , Enfermedad de Huntington/metabolismo , Inflamación/inmunología , Inflamación/metabolismo , Enfermedad de Parkinson/metabolismo , Enfermedades por Prión/enzimología , Enfermedades por Prión/metabolismo , Enfermedades por Prión/transmisión , Priones/genética , Priones/patogenicidad , Deficiencias en la Proteostasis/enzimología , Deficiencias en la Proteostasis/genética , Factores de Riesgo , Reacción a la Transfusión/epidemiología
2.
Mol Cell ; 48(5): 747-59, 2012 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-23123197

RESUMEN

NPGPx is a member of the glutathione peroxidase (GPx) family; however, it lacks GPx enzymatic activity due to the absence of a critical selenocysteine residue, rendering its function an enigma. Here, we show that NPGPx is a newly identified stress sensor that transmits oxidative stress signals by forming the disulfide bond between its Cys57 and Cys86 residues. This oxidized form of NPGPx binds to glucose-regulated protein (GRP)78 and forms covalent bonding intermediates between Cys86 of NPGPx and Cys41/Cys420 of GRP78. Subsequently, the formation of the disulfide bond between Cys41 and Cys420 of GRP78 enhances its chaperone activity. NPGPx-deficient cells display increased reactive oxygen species, accumulated misfolded proteins, and impaired GRP78 chaperone activity. Complete loss of NPGPx in animals causes systemic oxidative stress, increases carcinogenesis, and shortens life span. These results suggest that NPGPx is essential for releasing excessive ER stress by enhancing GRP78 chaperone activity to maintain physiological homeostasis.


Asunto(s)
Proteínas Portadoras/metabolismo , Estrés del Retículo Endoplásmico , Proteínas de Choque Térmico/metabolismo , Estrés Oxidativo , Peroxidasas/metabolismo , Deficiencias en la Proteostasis/enzimología , Transducción de Señal , Animales , Proteínas Portadoras/genética , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Cisteína , Daño del ADN , Disulfuros/metabolismo , Relación Dosis-Respuesta a Droga , Chaperón BiP del Retículo Endoplásmico , Estrés del Retículo Endoplásmico/efectos de los fármacos , Estrés del Retículo Endoplásmico/genética , Fibroblastos/enzimología , Fibroblastos/patología , Glutatión Peroxidasa , Proteínas de Choque Térmico/genética , Homeostasis , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutagénesis Sitio-Dirigida , Mutación , Oxidantes/farmacología , Oxidación-Reducción , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/genética , Peroxidasas/genética , Unión Proteica , Pliegue de Proteína , Deficiencias en la Proteostasis/genética , Deficiencias en la Proteostasis/patología , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Factores de Tiempo , Transfección
3.
J Biol Chem ; 292(45): 18392-18407, 2017 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-28939772

RESUMEN

p97 is an essential ATPase associated with various cellular activities (AAA+) that functions as a segregase in diverse cellular processes, including the maintenance of proteostasis. p97 interacts with different cofactors that target it to distinct pathways; an important example is the deubiquitinase ataxin3, which collaborates with p97 in endoplasmic reticulum-associated degradation. However, the molecular details of this interaction have been unclear. Here, we characterized the binding of ataxin3 to p97, showing that ataxin3 binds with low-micromolar affinity to both wild-type p97 and mutants linked to degenerative disorders known as multisystem proteinopathy 1 (MSP1); we further showed that the stoichiometry of binding is one ataxin3 molecule per p97 hexamer. We mapped the binding determinants on each protein, demonstrating that ataxin3's p97/VCP-binding motif interacts with the inter-lobe cleft in the N-domain of p97. We also probed the nucleotide dependence of this interaction, confirming that ataxin3 and p97 associate in the presence of ATP and in the absence of nucleotide, but not in the presence of ADP. Our experiments suggest that an ADP-driven downward movement of the p97 N-terminal domain dislodges ataxin3 by inducing a steric clash between the D1-domain and ataxin3's C terminus. In contrast, MSP1 mutants of p97 bind ataxin3 irrespective of their nucleotide state, indicating a failure by these mutants to translate ADP binding into a movement of the N-terminal domain. Our model provides a mechanistic explanation for how nucleotides regulate the p97-ataxin3 interaction and why atypical cofactor binding is observed with MSP1 mutants.


Asunto(s)
Ataxina-3/metabolismo , Coenzimas/metabolismo , Miopatías Distales/metabolismo , Modelos Moleculares , Deficiencias en la Proteostasis/metabolismo , Proteínas Represoras/metabolismo , Proteína que Contiene Valosina/metabolismo , Adenosina Difosfato/química , Adenosina Difosfato/metabolismo , Adenosina Trifosfato/química , Adenosina Trifosfato/metabolismo , Sustitución de Aminoácidos , Ataxina-3/química , Ataxina-3/genética , Sitios de Unión , Unión Competitiva , Coenzimas/química , Coenzimas/genética , Cristalografía por Rayos X , Bases de Datos de Proteínas , Miopatías Distales/enzimología , Miopatías Distales/genética , Humanos , Microscopía Electrónica de Transmisión , Mutación , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Mapeo de Interacción de Proteínas , Multimerización de Proteína , Deficiencias en la Proteostasis/enzimología , Deficiencias en la Proteostasis/genética , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Represoras/química , Proteínas Represoras/genética , Proteína que Contiene Valosina/química , Proteína que Contiene Valosina/genética
4.
Biochem J ; 474(14): 2333-2347, 2017 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-28673927

RESUMEN

Glycosylation constitutes one of the most common, ubiquitous and complex forms of post-translational modification. It commences with the synthesis of the protein and plays a significant role in deciding its folded state, oligomerization and thus its function. Recent studies have demonstrated that N-linked glycans help proteins to fold as the stability and folding kinetics are altered with the removal of the glycans from them. Several studies have shown that it alters not only the thermodynamic stability but also the structural features of the folded proteins modulating their interactions and functions. Their inhibition and perturbations have been implicated in diseases from diabetes to degenerative disorders. The intent of this review is to provide insight into the recent advancements in the general understanding on the aspect of glycosylation driven stability of proteins that is imperative to their function and finally their role in health and disease states.


Asunto(s)
Glicósido Hidrolasas/metabolismo , Glicosiltransferasas/metabolismo , Modelos Biológicos , Modelos Moleculares , Procesamiento Proteico-Postraduccional , Proteínas/metabolismo , Animales , Asparagina/metabolismo , Glicosilación , Humanos , Cinética , Conformación Proteica , Pliegue de Proteína , Multimerización de Proteína , Estabilidad Proteica , Proteínas/química , Deficiencias en la Proteostasis/enzimología , Deficiencias en la Proteostasis/metabolismo , Termodinámica
5.
J Biol Chem ; 291(41): 21363-21374, 2016 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-27551041

RESUMEN

Congenital insensitivity to pain with anhidrosis (CIPA) is a rare autosomal recessive disorder characterized by insensitivity to noxious stimuli and variable intellectual disability (ID) due to mutations in the NTRK1 gene encoding the NGF receptor TrkA. To get an insight in the effect of NTRK1 mutations in the cognitive phenotype we biochemically characterized three TrkA mutations identified in children diagnosed of CIPA with variable ID. These mutations are located in different domains of the protein; L213P in the extracellular domain, Δ736 in the kinase domain, and C300stop in the extracellular domain, a new mutation causing CIPA diagnosed in a Spanish teenager. We found that TrkA mutations induce misfolding, retention in the endoplasmic reticulum (ER), and aggregation in a mutation-dependent manner. The distinct mutations are degraded with a different kinetics by different ER quality control mechanisms; although C300stop is rapidly disposed by autophagy, Δ736 degradation is sensitive to the proteasome and to autophagy inhibitors, and L213P is a long-lived protein refractory to degradation. In addition L213P enhances the formation of autophagic vesicles triggering an increase in the autophagic flux with deleterious consequences. Mouse cortical neurons expressing L213P showed the accumulation of LC3-GFP positive puncta and dystrophic neurites. Our data suggest that TrkA misfolding and aggregation induced by some CIPA mutations disrupt the autophagy homeostasis causing neurodegeneration. We propose that distinct disease-causing mutations of TrkA generate different levels of cell toxicity, which may provide an explanation of the variable intellectual disability observed in CIPA patients.


Asunto(s)
Autofagia , Hipohidrosis/enzimología , Mutación Missense , Enfermedades Neurodegenerativas/enzimología , Insensibilidad Congénita al Dolor/enzimología , Agregación Patológica de Proteínas/enzimología , Deficiencias en la Proteostasis/enzimología , Receptor trkA/metabolismo , Adolescente , Sustitución de Aminoácidos , Animales , Corteza Cerebral/enzimología , Femenino , Células HeLa , Humanos , Hipohidrosis/genética , Masculino , Ratones , Ratones Mutantes , Enfermedades Neurodegenerativas/genética , Nociceptores/enzimología , Insensibilidad Congénita al Dolor/genética , Agregación Patológica de Proteínas/genética , Deficiencias en la Proteostasis/genética , Receptor trkA/genética
6.
J Inherit Metab Dis ; 38(2): 287-94, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25331909

RESUMEN

Classical homocystinuria is caused by mutations in the cystathionine ß-synthase (CBS) gene. Previous experiments in bacterial and yeast cells showed that many mutant CBS enzymes misfold and that chemical chaperones enable proper folding of a number of mutations. In the present study, we tested the extent of misfolding of 27 CBS mutations previously tested in E. coli under the more folding-permissive conditions of mammalian CHO-K1 cells and the ability of chaperones to rescue the conformation of these mutations. Expression of mutations in mammalian cells increased the median activity 16-fold and the amount of tetramers 3.2-fold compared with expression in bacteria. Subsequently, we tested the responses of seven selected mutations to three compounds with chaperone-like activity. Aminooxyacetic acid and 4-phenylbutyric acid exhibited only a weak effect. In contrast, heme arginate substantially increased the formation of mutant CBS protein tetramers (up to sixfold) and rescued catalytic activity (up to ninefold) of five out of seven mutations (p.A114V, p.K102N, p.R125Q, p.R266K, and p.R369C). The greatest effect of heme arginate was observed for the mutation p.R125Q, which is non-responsive to in vivo treatment with vitamin B(6). Moreover, the heme responsiveness of the p.R125Q mutation was confirmed in fibroblasts derived from a patient homozygous for this genetic variant. Based on these data, we propose that a distinct group of heme-responsive CBS mutations may exist and that the heme pocket of CBS may become an important target for designing novel therapies for homocystinuria.


Asunto(s)
Arginina/farmacología , Cistationina betasintasa/genética , Fibroblastos/efectos de los fármacos , Hemo/farmacología , Homocistinuria/tratamiento farmacológico , Chaperonas Moleculares/farmacología , Mutación , Deficiencias en la Proteostasis/tratamiento farmacológico , Animales , Células CHO , Dominio Catalítico , Cricetulus , Cistationina betasintasa/metabolismo , Femenino , Fibroblastos/enzimología , Predisposición Genética a la Enfermedad , Homocistinuria/diagnóstico , Homocistinuria/enzimología , Homocistinuria/genética , Homocigoto , Humanos , Modelos Moleculares , Fenotipo , Conformación Proteica , Pliegue de Proteína , Deficiencias en la Proteostasis/diagnóstico , Deficiencias en la Proteostasis/enzimología , Deficiencias en la Proteostasis/genética , Relación Estructura-Actividad , Especificidad por Sustrato , Transfección
7.
Biochim Biophys Acta ; 1832(8): 1279-93, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23583749

RESUMEN

Type I galactosemia is a genetic disorder that is caused by the impairment of galactose-1-phosphate uridylyltransferase (GALT; EC 2.7.7.12). Although a large number of mutations have been detected through genetic screening of the human GALT (hGALT) locus, for many it is not known how they cause their effects. The majority of these mutations are missense, with predicted substitutions scattered throughout the enzyme structure and thus causing impairment by other means rather than direct alterations to the active site. To clarify the fundamental, molecular basis of hGALT impairment we studied five disease-associated variants p.D28Y, p.L74P, p.F171S, p.F194L and p.R333G using both a yeast model and purified, recombinant proteins. In a yeast expression system there was a correlation between lysate activity and the ability to rescue growth in the presence of galactose, except for p.R333G. Kinetic analysis of the purified proteins quantified each variant's level of enzymatic impairment and demonstrated that this was largely due to altered substrate binding. Increased surface hydrophobicity, altered thermal stability and changes in proteolytic sensitivity were also detected. Our results demonstrate that hGALT requires a level of flexibility to function optimally and that altered folding is the underlying reason of impairment in all the variants tested here. This indicates that misfolding is a common, molecular basis of hGALT deficiency and suggests the potential of pharmacological chaperones and proteostasis regulators as novel therapeutic approaches for type I galactosemia.


Asunto(s)
Galactosemias/enzimología , Deficiencias en la Proteostasis/enzimología , UDP-Glucosa-Hexosa-1-Fosfato Uridiltransferasa/química , UDP-Glucosa-Hexosa-1-Fosfato Uridiltransferasa/metabolismo , Galactosemias/etiología , Galactosemias/genética , Humanos , Cinética , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Mutación , Unión Proteica , Desnaturalización Proteica , Deficiencias en la Proteostasis/etiología , Deficiencias en la Proteostasis/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , UDP-Glucosa-Hexosa-1-Fosfato Uridiltransferasa/genética
8.
Biochem J ; 452(3): 391-400, 2013 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-23566014

RESUMEN

Mutations in the CLCN5 (chloride channel, voltage-sensitive 5) gene cause Dent's disease because they reduce the functional expression of the ClC-5 chloride/proton transporter in the recycling endosomes of proximal tubule epithelial cells. The majority (60%) of these disease-causing mutations in ClC-5 are misprocessed and retained in the ER (endoplasmic reticulum). Importantly, the structural basis for misprocessing and the cellular destiny of such ClC-5 mutants have yet to be defined. A ClC-5 monomer comprises a short N-terminal region, an extensive membrane domain and a large C-terminal domain. The recent crystal structure of a eukaryotic ClC (chloride channel) transporter revealed the intimate interaction between the membrane domain and the C-terminal region. Therefore we hypothesized that intramolecular interactions may be perturbed in certain mutants. In the present study we examined two misprocessed mutants: C221R located in the membrane domain and R718X, which truncates the C-terminal domain. Both mutants exhibited enhanced protease susceptibility relative to the normal protein in limited proteolysis studies, providing direct evidence that they are misfolded. Interestingly, the membrane-localized mutation C221R led to enhanced protease susceptibility of the cytosolic N-terminal region, and the C-terminal truncation mutation R718X led to enhanced protease susceptibility of both the cytosolic C-terminal and the membrane domain. Together, these studies support the idea that certain misprocessing mutations alter intramolecular interactions within the full-length ClC-5 protein. Further, we found that these misfolded mutants are polyubiquitinated and targeted for proteasomal degradation in the OK (opossum kidney) renal epithelial cells, thereby ensuring that they do not elicit the unfolded protein response.


Asunto(s)
Canales de Cloruro/química , Canales de Cloruro/genética , Codón sin Sentido/genética , Enfermedad de Dent/genética , Mutación Missense/genética , Complejo de la Endopetidasa Proteasomal/química , Complejo de la Endopetidasa Proteasomal/genética , Animales , Enfermedad de Dent/enzimología , Enfermedad de Dent/metabolismo , Retículo Endoplásmico/química , Retículo Endoplásmico/genética , Células HEK293 , Humanos , Zarigüeyas , Complejo de la Endopetidasa Proteasomal/metabolismo , Conformación Proteica , Procesamiento Proteico-Postraduccional/genética , Deficiencias en la Proteostasis/enzimología , Deficiencias en la Proteostasis/genética , Deficiencias en la Proteostasis/metabolismo
9.
Artículo en Inglés | MEDLINE | ID: mdl-24814990

RESUMEN

Chaperone therapy is a newly developed molecular therapeutic approach to protein misfolding diseases. Among them we found unstable mutant enzyme proteins in a few lysosomal diseases, resulting in rapid intracellular degradation and loss of function. Active-site binding low molecular competitive inhibitors (chemical chaperones) paradoxically stabilized and enhanced the enzyme activity in somatic cells by correction of the misfolding of enzyme protein. They reached the brain through the blood-brain barrier after oral administration, and corrected pathophysiology of the disease. In addition to these inhibitory chaperones, non-competitive chaperones without inhibitory bioactivity are being developed. Furthermore molecular chaperone therapy utilizing the heat shock protein and other chaperone proteins induced by small molecules has been experimentally tried to handle abnormally accumulated proteins as a new approach particularly to neurodegenerative diseases. These three types of chaperones are promising candidates for various types of diseases, genetic or non-genetic, and neurological or non-neurological, in addition to lysosomal diseases.


Asunto(s)
Chaperonas Moleculares/metabolismo , Terapia Molecular Dirigida/métodos , Deficiencias en la Proteostasis/tratamiento farmacológico , Edad de Inicio , Animales , Humanos , Chaperonas Moleculares/genética , Mutación , Deficiencias en la Proteostasis/enzimología , Deficiencias en la Proteostasis/epidemiología , Deficiencias en la Proteostasis/genética
10.
Amino Acids ; 45(6): 1331-41, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24178766

RESUMEN

Many inborn errors of amino acids metabolism are caused by single point mutations affecting the ability of proteins to fold properly (i.e., protein homeostasis), thus leading to enzyme loss-of-function. Mutations may affect protein homeostasis by altering intrinsic physical properties of the polypeptide (folding thermodynamics, and rates of folding/unfolding/misfolding) as well as the interaction of partially folded states with elements of the protein homeostasis network (such as molecular chaperones and proteolytic machineries). Understanding these mutational effects on protein homeostasis is required to develop new therapeutic strategies aimed to target specific features of the mutant polypeptide. Here, I review recent work in three different diseases of protein homeostasis associated to inborn errors of amino acids metabolism: phenylketonuria, inherited homocystinuria and primary hyperoxaluria type I. These three different genetic disorders involve proteins operating in different cell organelles and displaying different structural complexities. Mutations often decrease protein kinetic stability of the native state (i.e., its half-life for irreversible denaturation), which can be studied using simple kinetic models amenable to biophysical and biochemical characterization. Natural ligands and pharmacological chaperones are shown to stabilize mutant enzymes, thus supporting their therapeutic application to overcome protein kinetic destabilization. The role of molecular chaperones in protein folding and misfolding is also discussed as well as their potential pharmacological modulation as promising new therapeutic approaches. Since current available treatments for these diseases are either burdening or only successful in a fraction of patients, alternative treatments must be considered covering studies from protein structure and biophysics to studies in animal models and patients.


Asunto(s)
Aminoácidos/metabolismo , Homeostasis , Estabilidad Proteica , Proteínas/metabolismo , Deficiencias en la Proteostasis/enzimología , Deficiencias en la Proteostasis/metabolismo , Animales , Humanos , Cinética , Mutación Puntual , Pliegue de Proteína , Proteínas/química , Deficiencias en la Proteostasis/genética , Deficiencias en la Proteostasis/terapia
11.
Physiol Rep ; 7(3): e13979, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30740912

RESUMEN

Homozygous mutations in NGLY1 were recently found to cause a condition characterized by a complex neurological syndrome, hypo- or alacrimia, and elevated liver transaminases. For yet unknown reasons, mortality is increased in patients with this condition. NGLY1 encodes the cytosolic enzyme N-glycanase 1, which is responsible for the deglycosylation of misfolded N-glycosylated proteins. Disruption of this process is hypothesized to lead to an accumulation of misfolded proteins in the cytosol. Here, we describe the disease course of a girl with a homozygous mutation in NGLY1, namely c.1837del (p.Gln613 fs). In addition to the previously described symptoms, at the age of 8 she presented with recurrent infections and hyperpigmentation, and, subsequently, a diagnosis of primary adrenal insufficiency was made. There are no previous reports describing adrenal insufficiency in such patients. We postulate that patients with NGLY1 deficiency may develop adrenal insufficiency as a consequence of impaired proteostasis, and the accompanying proteotoxic stress-induced cell death, through defective Nrf1 function. We recommend an annual evaluation of adrenal function in all patients with NGLY1 mutations in order to prevent unnecessary deaths.


Asunto(s)
Insuficiencia Suprarrenal/genética , Trastornos Congénitos de Glicosilación/genética , Mutación , Péptido-N4-(N-acetil-beta-glucosaminil) Asparagina Amidasa/deficiencia , Deficiencias en la Proteostasis/genética , Proteostasis/genética , Insuficiencia Suprarrenal/diagnóstico , Insuficiencia Suprarrenal/enzimología , Niño , Trastornos Congénitos de Glicosilación/complicaciones , Trastornos Congénitos de Glicosilación/diagnóstico , Trastornos Congénitos de Glicosilación/enzimología , Femenino , Predisposición Genética a la Enfermedad , Homocigoto , Humanos , Péptido-N4-(N-acetil-beta-glucosaminil) Asparagina Amidasa/genética , Fenotipo , Pronóstico , Deficiencias en la Proteostasis/diagnóstico , Deficiencias en la Proteostasis/enzimología
12.
Mol Neurodegener ; 12(1): 2, 2017 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-28057013

RESUMEN

BACKGROUND: Dominant mutations in Cu/Zn-superoxide dismutase (SOD1) gene cause a familial form of amyotrophic lateral sclerosis (SOD1-ALS) with accumulation of misfolded SOD1 proteins as intracellular inclusions in spinal motor neurons. Oligomerization of SOD1 via abnormal disulfide crosslinks has been proposed as one of the misfolding pathways occurring in mutant SOD1; however, the pathological relevance of such oligomerization in the SOD1-ALS cases still remains obscure. METHODS: We prepared antibodies exclusively recognizing the SOD1 oligomers cross-linked via disulfide bonds in vitro. By using those antibodies, immunohistochemical examination and ELISA were mainly performed on the tissue samples of transgenic mice expressing mutant SOD1 proteins and also of human SOD1-ALS cases. RESULTS: We showed the recognition specificity of our antibodies exclusively toward the disulfide-crosslinked SOD1 oligomers by ELISA using various forms of purified SOD1 proteins in conformationally distinct states in vitro. Furthermore, the epitope of those antibodies was buried and inaccessible in the natively folded structure of SOD1. The antibodies were then found to specifically detect the pathological SOD1 species in the spinal motor neurons of the SOD1-ALS patients as well as the transgenic model mice. CONCLUSIONS: Our findings here suggest that the SOD1 oligomerization through the disulfide-crosslinking associates with exposure of the SOD1 structural interior and is a pathological process occurring in the SOD1-ALS cases.


Asunto(s)
Esclerosis Amiotrófica Lateral/enzimología , Superóxido Dismutasa-1/química , Superóxido Dismutasa-1/metabolismo , Animales , Western Blotting , Ensayo de Inmunoadsorción Enzimática , Humanos , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Deficiencias en la Proteostasis/enzimología
13.
Methods Mol Biol ; 1133: 223-35, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24567105

RESUMEN

The functional versatility of metacaspase proteases has been established by reports of their involvement in non-apoptotic cellular processes, in addition to their canonical role in apoptosis/programmed cell death. While the budding yeast metacaspase Yca1 has been well characterized for its role in cell death regulation, more recent examinations suggest that the protease may be involved in key processes that increase survival and fitness. More specifically, examinations suggest that Yca1 is central to maintaining cellular proteostasis as it interacts with major components involved in protein biosynthesis and functions to limit aggregate deposition. Here, we describe the methods utilized to analyze the role Yca1 in proteostasis.


Asunto(s)
Caspasas/biosíntesis , Biología Molecular/métodos , Proteínas de Saccharomyces cerevisiae/biosíntesis , Saccharomyces cerevisiae/enzimología , Apoptosis/genética , Caspasas/genética , Biosíntesis de Proteínas/genética , Deficiencias en la Proteostasis/enzimología , Deficiencias en la Proteostasis/genética , Proteínas de Saccharomyces cerevisiae/genética
14.
Clin Biochem ; 47(3): 158-65, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24355692

RESUMEN

The biologically active form of the B6 vitamers is pyridoxal 5'-phosphate (PLP), which plays a coenzymatic role in several distinct enzymatic activities ranging from the synthesis, interconversion and degradation of amino acids to the replenishment of one-carbon units, synthesis and degradation of biogenic amines, synthesis of tetrapyrrolic compounds and metabolism of amino-sugars. In the catalytic process of PLP-dependent enzymes, the substrate amino acid forms a Schiff base with PLP and the electrophilicity of the PLP pyridine ring plays important roles in the subsequent catalytic steps. While the essential role of PLP in the acquisition of biological activity of many proteins is long recognized, the finding that some PLP-enzymes require the coenzyme for refolding in vitro points to an additional role of PLP as a chaperone in the folding process. Mutations in the genes encoding PLP-enzymes are causative of several rare inherited diseases. Patients affected by some of these diseases (AADC deficiency, cystathionuria, homocystinuria, gyrate atrophy, primary hyperoxaluria type 1, xanthurenic aciduria, X-linked sideroblastic anaemia) can benefit, although at different degrees, from the administration of pyridoxine, a PLP precursor. The effect of the coenzyme is not limited to mutations that affect the enzyme-coenzyme interaction, but also to those that cause folding defects, reinforcing the idea that PLP could play a chaperone role and improve the folding efficiency of misfolded variants. In this review, recent biochemical and cell biology studies highlighting the chaperoning activity of the coenzyme on folding-defective variants of PLP-enzymes associated with rare diseases are presented and discussed.


Asunto(s)
Coenzimas/metabolismo , Errores Innatos del Metabolismo/enzimología , Chaperonas Moleculares/metabolismo , Fosfato de Piridoxal/metabolismo , Vitamina B 6/metabolismo , Animales , Coenzimas/genética , Humanos , Errores Innatos del Metabolismo/genética , Chaperonas Moleculares/genética , Pliegue de Proteína , Deficiencias en la Proteostasis/enzimología , Deficiencias en la Proteostasis/genética , Fosfato de Piridoxal/genética , Vitamina B 6/genética
15.
Mol Cell Endocrinol ; 382(1): 55-65, 2014 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-24001579

RESUMEN

GCK-MODY, dominantly inherited mild hyperglycemia, is associated with more than 600 mutations in the glucokinase gene. Different molecular mechanisms have been shown to explain GCK-MODY. Here, we report a Pakistani family harboring the glucokinase mutation c.823C>T (p.R275C). The recombinant and in cellulo expressed mutant pancreatic enzyme revealed slightly increased enzyme activity (kcat) and normal affinity for α-D-glucose, and resistance to limited proteolysis by trypsin comparable with wild-type. When stably expressed in HEK293 cells and MIN6 ß-cells (at different levels), the mutant protein appeared misfolded and unstable with a propensity to form dimers and aggregates. Its degradation rate was increased, involving the lysosomal and proteasomal quality control systems. On mutation, a hydrogen bond between the R275 side-chain and the carbonyl oxygen of D267 is broken, destabilizing the F260-L271 loop structure and the protein. This promotes the formation of dimers/aggregates and suggests that an increased cellular degradation is the molecular mechanism by which R275C causes GCK-MODY.


Asunto(s)
Diabetes Mellitus Tipo 2/enzimología , Glucoquinasa/genética , Proteínas Mutantes/metabolismo , Mutación/genética , Pliegue de Proteína , Proteolisis , Deficiencias en la Proteostasis/enzimología , Adulto , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/patología , Estabilidad de Enzimas , Familia , Femenino , Glucoquinasa/química , Glucoquinasa/metabolismo , Glucosa/metabolismo , Células HEK293 , Humanos , Cinética , Lisosomas/metabolismo , Masculino , Proteínas Mutantes/química , Pakistán , Linaje , Fenotipo , Complejo de la Endopetidasa Proteasomal/metabolismo , Multimerización de Proteína , Deficiencias en la Proteostasis/genética , Deficiencias en la Proteostasis/patología , Proteínas Recombinantes/metabolismo
16.
FEBS Lett ; 587(13): 1981-7, 2013 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-23684649

RESUMEN

Stress-denatured or de novo synthesized and translocated unfolded polypeptides can spontaneously reach their native state without assistance of other proteins. Yet, the pathway to native folding is complex, stress-sensitive and prone to errors. Toxic misfolded and aggregated conformers may accumulate in cells and lead to degenerative diseases. Members of the canonical conserved families of molecular chaperones, Hsp100s, Hsp70/110/40s, Hsp60/CCTs, the small Hsps and probably also Hsp90s, can recognize and bind with high affinity, abnormally exposed hydrophobic surfaces on misfolded and aggregated polypeptides. Binding to Hsp100, Hsp70, Hsp110, Hsp40, Hsp60, CCTs and Trigger factor may cause partial unfolding of the misfolded polypeptide substrates, and ATP hydrolysis can induce further unfolding and release from the chaperone, leading to spontaneous refolding into native proteins with low-affinity for the chaperones. Hence, specific chaperones act as catalytic polypeptide unfolding isomerases, rerouting cytotoxic misfolded and aggregated polypeptides back onto their physiological native refolding pathway, thus averting the onset of protein conformational diseases.


Asunto(s)
Chaperoninas/fisiología , Péptidos/metabolismo , Desplegamiento Proteico , Animales , Biocatálisis , Proteínas de Choque Térmico/fisiología , Humanos , Deficiencias en la Proteostasis/enzimología
17.
Curr Opin Cell Biol ; 23(2): 223-30, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21295458

RESUMEN

A hallmark event in neurodegenerative diseases is the accumulation of misfolded aggregated proteins in the brain leading to neuronal dysfunction and disease. Compelling evidence suggests that misfolded proteins damage cells by inducing endoplasmic reticulum (ER) stress and alterations in calcium homeostasis. Changes in cytoplasmic calcium concentration lead to unbalances on several signaling pathways. Recent data suggest that calcium-mediated hyperactivation of calcineurin (CaN), a key phosphatase in the brain, triggers synaptic dysfunction and neuronal death, the two central events responsible for brain degeneration in neurodegenerative diseases. Therefore, blocking CaN hyper-activation might be a promising therapeutic strategy to prevent brain damage in neurodegenerative diseases.


Asunto(s)
Calcineurina/metabolismo , Retículo Endoplásmico/enzimología , Enfermedades Neurodegenerativas/enzimología , Deficiencias en la Proteostasis/enzimología , Estrés Fisiológico , Animales , Humanos
18.
Antioxid Redox Signal ; 14(8): 1479-92, 2011 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-20812868

RESUMEN

Excessive nitrosative and oxidative stress is thought to trigger cellular signaling pathways leading to neurodegenerative conditions. Such redox dysregulation can result from many cellular events, including hyperactivation of the N-methyl-D-aspartate-type glutamate receptor, mitochondrial dysfunction, and cellular aging. Recently, we and our colleagues have shown that excessive generation of free radicals and related molecules, in particular nitric oxide species (NO), can trigger pathological production of misfolded proteins, abnormal mitochondrial dynamics (comprised of mitochondrial fission and fusion events), and apoptotic pathways in neuronal cells. Emerging evidence suggests that excessive NO production can contribute to these pathological processes, specifically by S-nitrosylation of specific target proteins. Here, we highlight examples of S-nitrosylated proteins that regulate misfolded protein accumulation and mitochondrial dynamics. For instance, in models of Parkinson's disease, these S-nitrosylation targets include parkin, a ubiquitin E3 ligase and neuroprotective molecule, and protein-disulfide isomerase, a chaperone enzyme for nascent protein folding. S-Nitrosylation of protein-disulfide isomerase may also be associated with mutant Cu/Zn superoxide dismutase toxicity in amyotrophic lateral sclerosis. Additionally, in models of Alzheimer's disease, excessive NO generation leads to the formation of S-nitrosylated dynamin-related protein 1 (forming SNO-Drp1), which contributes to abnormal mitochondrial fragmentation and resultant synaptic damage.


Asunto(s)
Mitocondrias/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Óxido Nítrico/metabolismo , Proteína Disulfuro Isomerasas/metabolismo , Deficiencias en la Proteostasis/metabolismo , Compuestos de Sulfhidrilo/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Humanos , Mitocondrias/patología , Enfermedades Neurodegenerativas/enzimología , Enfermedades Neurodegenerativas/patología , Pliegue de Proteína , Deficiencias en la Proteostasis/enzimología , Deficiencias en la Proteostasis/patología
19.
Ageing Res Rev ; 10(4): 498-502, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21651995

RESUMEN

Research carried out up to 3 decades ago by Gracy and co-workers revealed that the activity of the glycolytic enzyme triosephosphate isomerase (TPI), which converts dihydroxyacetone phosphate (DHAP) to glyceraldehyde-3-phosphate (G3P), gradually declines whilst carrying out its catalytic function, primarily due to deamidation of certain asparagine residues. It is suggested here that excessive or continuous glycolysis increases TPI deamidation and thereby lowers TPI activity and causes accumulation of its substrate, DHAP, which in turn decomposes into methylglyoxal (MG), a well-recognised reactive bicarbonyl whose actions in cells and tissues, as well as at the whole organism level, mimic much age-relate dysfunction. The proposal helps to explain why suppression of glycolysis by caloric restriction, fasting and increased aerobic activity also suppresses generation of altered proteins which characterise the aged phenotype. It is proposed that these effects on TPI activity, though seemingly neglected in biogerontological contexts, reveal a mechanistic link between energy metabolism and age-related proteostatic dysfunction.


Asunto(s)
Senescencia Celular , Metabolismo Energético/fisiología , Deficiencias en la Proteostasis/metabolismo , Triosa-Fosfato Isomerasa/metabolismo , Animales , Senescencia Celular/genética , Senescencia Celular/fisiología , Metabolismo Energético/genética , Glucólisis/genética , Glucólisis/fisiología , Humanos , Fenotipo , Deficiencias en la Proteostasis/enzimología , Deficiencias en la Proteostasis/patología , Distribución Tisular/genética , Distribución Tisular/fisiología , Triosa-Fosfato Isomerasa/genética
20.
Trends Cardiovasc Med ; 21(8): 224-8, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22902070

RESUMEN

As exemplified by desmin-related cardiomyopathy and myocardial ischemia/reperfusion injury, proteasome functional insufficiency plays an essential pathogenic role in the progression of cardiac diseases with elevated proteotoxic stress. Upregulation of p62/SQSTM1 and increased selective autophagy in cardiomyocytes may protect against proteotoxic stress in the heart. p62 may serve as a proteotoxic stress sensor, promote segregation and degradation of misfolded proteins by autophagy, and mediate the cross talk between the ubiquitin-proteasome system and autophagy.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Autofagia , Cardiopatías/enzimología , Miocardio/enzimología , Complejo de la Endopetidasa Proteasomal/metabolismo , Deficiencias en la Proteostasis/enzimología , Transducción de Señal , Estrés Fisiológico , Ubiquitina/metabolismo , Animales , Cardiopatías/patología , Humanos , Miocardio/patología , Pliegue de Proteína , Deficiencias en la Proteostasis/patología , Proteína Sequestosoma-1 , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA