Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 156
Filtrar
Más filtros

Medicinas Complementárias
Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Arch Biochem Biophys ; 716: 109112, 2022 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-34954215

RESUMEN

In apoptotic pathway, the interaction of Cytochrome c (Cytc) with cardiolipin in vivo is a key process to induce peroxidase activity of Cytc and trigger the release of Cytc in the inner mitochondria into cytosol. The peroxidase active form of Cytc occurs due to local conformational changes that support the opening of the heme crevice and the loss of an axial ligand between Met80 and heme Fe. Structural adjustments at the Ω-loop segments of Cytc are required for such process. To study the role of the distal Ω-loop segments comprising residues 71-85 in human Cytc (hCytc), we investigated a cysteine mutation at Pro76, one of the highly conserved residues in this loop. The effect of P76C mutant was explored by the combination of experimental characterizations and molecular dynamics (MD) simulations. The peroxidase activity of the P76C mutant was found to be significantly increased by ∼13 folds relative to the wild type. Experimental data on global denaturation, alkaline transition, heme bleaching, and spin-labeling Electron Spin Resonance were in good agreement with the enhancement of peroxidase activity. The MD results of hCytc in the hexacoordinate form suggest the important changes in P76C mutant occurred due to the unfolding at the central Ω-loop (residues 40-57), and the weakening of H-bond between Tyr67 and Met80. Whereas the experimental data implied that the P76C mutant tend to be in equilibrium between the pentacoordinate and hexacoordinate forms, the MD and experimental information are complementary and were used to support the mechanisms of peroxidase active form of hCytc.


Asunto(s)
Citocromos c/metabolismo , Proteínas Mutantes/metabolismo , Peroxidasas/metabolismo , Secuencia de Aminoácidos , Cardiolipinas/metabolismo , Cisteína/química , Citocromos c/genética , Activación Enzimática , Hemo/metabolismo , Humanos , Simulación de Dinámica Molecular , Proteínas Mutantes/genética , Mutación , Conformación Proteica , Relación Estructura-Actividad
2.
J Clin Invest ; 131(6)2021 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-33507883

RESUMEN

Omega-3 fatty acids from fish oil reduce triglyceride levels in mammals, yet the mechanisms underlying this effect have not been fully clarified, despite the clinical use of omega-3 ethyl esters to treat severe hypertriglyceridemia and reduce cardiovascular disease risk in humans. Here, we identified in bile a class of hypotriglyceridemic omega-3 fatty acid-derived N-acyl taurines (NATs) that, after dietary omega-3 fatty acid supplementation, increased to concentrations similar to those of steroidal bile acids. The biliary docosahexaenoic acid-containing (DHA-containing) NAT C22:6 NAT was increased in human and mouse plasma after dietary omega-3 fatty acid supplementation and potently inhibited intestinal triacylglycerol hydrolysis and lipid absorption. Supporting this observation, genetic elevation of endogenous NAT levels in mice impaired lipid absorption, whereas selective augmentation of C22:6 NAT levels protected against hypertriglyceridemia and fatty liver. When administered pharmacologically, C22:6 NAT accumulated in bile and reduced high-fat diet-induced, but not sucrose-induced, hepatic lipid accumulation in mice, suggesting that C22:6 NAT is a negative feedback mediator that limits excess intestinal lipid absorption. Thus, biliary omega-3 NATs may contribute to the hypotriglyceridemic mechanism of action of fish oil and could influence the design of more potent omega-3 fatty acid-based therapeutics.


Asunto(s)
Ácidos Grasos Omega-3/administración & dosificación , Hipertrigliceridemia/dietoterapia , Triglicéridos/metabolismo , Amidohidrolasas/deficiencia , Amidohidrolasas/genética , Amidohidrolasas/metabolismo , Animales , Bilis/metabolismo , Modelos Animales de Enfermedad , Ácidos Docosahexaenoicos/análogos & derivados , Ácidos Docosahexaenoicos/metabolismo , Ácidos Grasos Omega-3/metabolismo , Hígado Graso/metabolismo , Hígado Graso/prevención & control , Humanos , Hipertrigliceridemia/metabolismo , Hipolipemiantes/administración & dosificación , Hipolipemiantes/metabolismo , Absorción Intestinal/efectos de los fármacos , Metabolismo de los Lípidos , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mutación Puntual , Taurina/análogos & derivados , Taurina/metabolismo
3.
J Mol Cell Cardiol ; 150: 77-90, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33148509

RESUMEN

BACKGROUND: The clinical outcome of hypertrophic cardiomyopathy patients is not only determined by the disease-causing mutation but influenced by a variety of disease modifiers. Here, we defined the role of the mutation location and the mutant protein dose of the troponin T mutations I79N, R94C and R278C. METHODS AND RESULTS: We determined myofilament function after troponin exchange in permeabilized single human cardiomyocytes as well as in cardiac patient samples harboring the R278C mutation. Notably, we found that a small dose of mutant protein is sufficient for the maximal effect on myofilament Ca2+-sensitivity for the I79N and R94C mutation while the mutation location determines the magnitude of this effect. While incorporation of I79N and R94C increased myofilament Ca2+-sensitivity, incorporation of R278C increased Ca2+-sensitivity at low and intermediate dose, while it decreased Ca2+-sensitivity at high dose. All three cTnT mutants showed reduced thin filament binding affinity, which coincided with a relatively low maximal exchange (50.5 ± 5.2%) of mutant troponin complex in cardiomyocytes. In accordance, 32.2 ± 4.0% mutant R278C was found in two patient samples which showed 50.0 ± 3.7% mutant mRNA. In accordance with studies that showed clinical variability in patients with the exact same mutation, we observed variability on the functional single cell level in patients with the R278C mutation. These differences in myofilament properties could not be explained by differences in the amount of mutant protein. CONCLUSIONS: Using troponin exchange in single human cardiomyocytes, we show that TNNT2 mutation-induced changes in myofilament Ca2+-sensitivity depend on mutation location, while all mutants show reduced thin filament binding affinity. The specific mutation-effect observed for R278C could not be translated to myofilament function of cardiomyocytes from patients, and is most likely explained by other (post)-translational troponin modifications. Overall, our studies illustrate that mutation location underlies variability in myofilament Ca2+-sensitivity, while only the R278C mutation shows a highly dose-dependent effect on myofilament function.


Asunto(s)
Cardiomiopatía Hipertrófica/genética , Cardiomiopatía Hipertrófica/patología , Mutación/genética , Miocitos Cardíacos/patología , Miofibrillas/patología , Troponina T/genética , Adolescente , Adulto , Anciano , Calcio/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Proteínas Mutantes/metabolismo , Miocitos Cardíacos/metabolismo , Miofibrillas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo
4.
Molecules ; 25(22)2020 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-33207644

RESUMEN

Functional nutrition is a valuable supplementation to dietary therapy. Functional foods are enriched with biologically active substances. Plant polyphenols attract particular attention due to multiple beneficial properties attributed to their high antioxidant and other biological activities. We assessed the effect of grape polyphenols on the life span of C57BL/6 mice and on behavioral and neuroinflammatory alterations in a transgenic mouse model of Parkinson disease (PD) with overexpression of the A53T-mutant human α-synuclein. C57BL/6 mice were given a dietary supplement containing grape polyphenol concentrate (GPC-1.5 mL/kg/day) with drinking water from the age of 6-8 weeks for life. Transgenic PD mice received GPC beginning at the age of 10 weeks for four months. GPC significantly influenced the cumulative proportion of surviving and substantially augmented the average life span in mice. In the transgenic PD model, the grape polyphenol (GP) diet enhanced memory reconsolidation and diminished memory extinction in a passive avoidance test. Behavioral effects of GP treatment were accompanied by a decrease in α-synuclein accumulation in the frontal cortex and a reduction in the expression of neuroinflammatory markers (IBA1 and CD54) in the frontal cortex and hippocampus. Thus, a GP-rich diet is recommended as promising functional nutrition for aging people and patients with neurodegenerative disorders.


Asunto(s)
Encéfalo/patología , Inflamación/tratamiento farmacológico , Degeneración Nerviosa/tratamiento farmacológico , Enfermedad de Parkinson/tratamiento farmacológico , Polifenoles/uso terapéutico , Vitis/química , Animales , Conducta Animal/efectos de los fármacos , Suplementos Dietéticos , Inflamación/complicaciones , Inflamación/patología , Ratones Endogámicos C57BL , Proteínas Mutantes/metabolismo , Degeneración Nerviosa/complicaciones , Enfermedad de Parkinson/complicaciones , Polifenoles/farmacología , Aumento de Peso/efectos de los fármacos , alfa-Sinucleína/metabolismo
5.
Artículo en Inglés | MEDLINE | ID: mdl-32296386

RESUMEN

Orexin receptors (OXRs) play a critical regulatory role in central control of food intake, maintenance of sleeping states, energy metabolism, and neuroendocrine homeostasis. However, most previous studies have focused on the sleep-promoting functions of OXRs in human beings, while their potential value in enhancing food intake for livestock breeding has not been fully exploited. In this study, we successfully cloned porcine orexin 2 receptor (pOX2R) complementary DNA and constructed four pOX2R mutants (P10S, P11T, V308I, and T401I) by site-directed mutagenesis, and their functional expressions were further confirmed through Western blotting analysis. Pharmacological characteristics of pOX2R and their mutants were further investigated. These results showed that the P10S, P11T, and T401I mutants had decreased cAMP signaling with orexin A, whereas only the P11T mutant decreased under the stimulation of orexin B. Besides, only P10S displayed a decreased calcium release in response to both orexin ligands. Importantly, these mutants exhibited decreased phosphorylation levels of ERK1/2, p38, and CREB to some degree compared with wild-type pOX2R. Collectively, these findings highlight the critical role of these mutations in pOX2R signaling and expand our understanding of molecular and pharmacological characterization of pOX2R.


Asunto(s)
Receptores de Orexina/metabolismo , Orexinas/farmacología , Porcinos , Animales , Clonación Molecular , Células HEK293 , Humanos , Modelos Moleculares , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mutación , Receptores de Orexina/química , Receptores de Orexina/genética , Orexinas/metabolismo , Filogenia , Conformación Proteica , Transducción de Señal/efectos de los fármacos , Porcinos/genética , Porcinos/metabolismo
6.
Commun Biol ; 3(1): 97, 2020 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-32139772

RESUMEN

Mutations to the gene encoding superoxide dismutase-1 (SOD1) were the first genetic elements discovered that cause motor neuron disease (MND). These mutations result in compromised SOD1 dimer stability, with one of the severest and most common mutations Ala4Val (A4V) displaying a propensity to monomerise and aggregate leading to neuronal death. We show that the clinically used ebselen and related analogues promote thermal stability of A4V SOD1 when binding to Cys111 only. We have developed a A4V SOD1 differential scanning fluorescence-based assay on a C6S mutation background that is effective in assessing suitability of compounds. Crystallographic data show that the selenium atom of these compounds binds covalently to A4V SOD1 at Cys111 at the dimer interface, resulting in stabilisation. This together with chemical amenability for hit expansion of ebselen and its on-target SOD1 pharmacological chaperone activity holds remarkable promise for structure-based therapeutics for MND using ebselen as a template.


Asunto(s)
Azoles/química , Azoles/farmacología , Diseño de Fármacos , Enfermedad de la Neurona Motora/tratamiento farmacológico , Compuestos de Organoselenio/química , Compuestos de Organoselenio/farmacología , Superóxido Dismutasa-1 , Sustitución de Aminoácidos/genética , Azoles/síntesis química , Azoles/uso terapéutico , Cristalografía por Rayos X , Descubrimiento de Drogas/métodos , Evaluación Preclínica de Medicamentos/métodos , Humanos , Isoindoles , Modelos Moleculares , Chaperonas Moleculares/síntesis química , Chaperonas Moleculares/química , Chaperonas Moleculares/uso terapéutico , Simulación del Acoplamiento Molecular , Enfermedad de la Neurona Motora/genética , Enfermedad de la Neurona Motora/metabolismo , Enfermedad de la Neurona Motora/patología , Proteínas Mutantes/química , Proteínas Mutantes/efectos de los fármacos , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mutación Missense , Compuestos de Organoselenio/síntesis química , Compuestos de Organoselenio/aislamiento & purificación , Compuestos de Organoselenio/uso terapéutico , Pliegue de Proteína/efectos de los fármacos , Multimerización de Proteína/efectos de los fármacos , Estabilidad Proteica/efectos de los fármacos , Estructura Terciaria de Proteína , Compuestos de Azufre/síntesis química , Compuestos de Azufre/química , Superóxido Dismutasa-1/química , Superóxido Dismutasa-1/efectos de los fármacos , Superóxido Dismutasa-1/genética , Superóxido Dismutasa-1/metabolismo , Termodinámica
7.
FEBS Lett ; 594(3): 424-438, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31605637

RESUMEN

NAD(P)H:quinone oxidoreductase 1 (NQO1) is a human FAD-dependent enzyme that plays a crucial role in the antioxidant defense system. A naturally occurring single-nucleotide polymorphism (NQO1*2) in the NQO1 gene leads to an amino acid substitution (P187S), which severely compromises the activity and stability of the enzyme. The NQO1*2 genotype has been linked to a higher risk for several types of cancer and poor survival rate after anthracycline-based chemotherapy. In this study, we show that a small molecular chaperone (N-(2-bromophenyl)pyrrolidine-1-sulfonamide) repopulates the native wild-type conformation. As a consequence of the stabilizing effect, the enzymatic activity of the P187S variant protein is strongly improved in the presence of the molecular chaperone in vitro.


Asunto(s)
Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mutación , NAD(P)H Deshidrogenasa (Quinona)/genética , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , Neoplasias/genética , Secuencia de Aminoácidos , Evaluación Preclínica de Medicamentos , Activación Enzimática/efectos de los fármacos , Estabilidad de Enzimas/efectos de los fármacos , Ligandos , Simulación del Acoplamiento Molecular , Proteínas Mutantes/antagonistas & inhibidores , Proteínas Mutantes/química , NAD(P)H Deshidrogenasa (Quinona)/antagonistas & inhibidores , NAD(P)H Deshidrogenasa (Quinona)/química , Conformación Proteica
8.
J Plant Physiol ; 245: 153091, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31887556

RESUMEN

Potato (Solanum tuberosum) mutant (ST) lacking one isoform of manganese-stabilizing protein (MSPI) of photosystem II exhibited besides spontaneous tuberization also growth changes with strongly impaired root system development. Previous studies revealed marked changes in carbohydrate levels and allocation within ST plant body. To verify causal relationship between changed carbohydrate balance and root growth restriction we engaged dark grown sucrose-supplied root organ-cultures of ST plants to exclude/confirm shoot effects. Unexpectedly, in ST root cultures we observed large alterations in growth and architecture as well as saccharide status similar to those found in the intact plant roots. The gene expression analysis, however, proved PsbO1 transcript (coding MSPI protein) neither in ST nor in WT root-organ cultures. Therefore, the results point to indirect effects of PsbO1 allele absence connected possibly with some epigenetic modulations.


Asunto(s)
Fotosíntesis/genética , Complejo de Proteína del Fotosistema II/metabolismo , Proteínas de Plantas/genética , Raíces de Plantas/metabolismo , Solanum tuberosum/genética , Alelos , Metabolismo de los Hidratos de Carbono/genética , Células Cultivadas , Regulación de la Expresión Génica de las Plantas/genética , Regulación de la Expresión Génica de las Plantas/fisiología , Manganeso/metabolismo , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mutación , Fotosíntesis/efectos de la radiación , Complejo de Proteína del Fotosistema II/genética , Proteínas de Plantas/metabolismo , Raíces de Plantas/crecimiento & desarrollo , Tubérculos de la Planta/genética , Tubérculos de la Planta/crecimiento & desarrollo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Solanum tuberosum/crecimiento & desarrollo , Sacarosa/metabolismo
9.
J Gerontol A Biol Sci Med Sci ; 75(4): 631-639, 2020 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-30346494

RESUMEN

Photobiomodulation using low-level light-emitting diode can be rapidly applied in neurological and physiological disorders safely and noninvasively. Photobiomodulation is effective for chronic diseases because of fewer side effects than drugs. Here we investigated the effects of photobiomodulation using light-emitting diode on amyloid plaques, gliosis, and neuronal loss to prevent and/or recover cognitive impairment, and optimal timing of photobiomodulation initiation for recovering cognitive function in a mouse model of Alzheimer's disease. 5XFAD mice were used as an Alzheimer's disease model. Animals receiving photobiomodulation treatment were divided into two groups: an early group starting photobiomodulation at 2 months of age (5XFAD+Early), and a late group starting photobiomodulation at 6 months of age (5XFAD+Delay). Both groups received photobiomodulation 20 minutes per session three times per week for 14 weeks. The Morris water maze, passive avoidance, and elevated plus maze tests were performed at 10 months of age. Immunohistochemistry and Western blot were performed after behavioral evaluation. The results showed that photobiomodulation treatment at early stages reduced amyloid accumulation, neuronal loss, and microgliosis and alleviated the cognitive dysfunction in 5XFAD mice, possibly by increasing insulin degrading enzyme related to amyloid-beta degradation. Photobiomodulation may be an excellent candidate for advanced preclinical Alzheimer's disease research.


Asunto(s)
Enfermedad de Alzheimer/radioterapia , Terapia por Luz de Baja Intensidad , Factores de Edad , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/psicología , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Reacción de Prevención/efectos de la radiación , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Corteza Cerebral/efectos de la radiación , Cognición/efectos de la radiación , Modelos Animales de Enfermedad , Gliosis/patología , Gliosis/prevención & control , Humanos , Láseres de Semiconductores/uso terapéutico , Masculino , Aprendizaje por Laberinto/efectos de la radiación , Ratones , Ratones Transgénicos , Microglía/metabolismo , Microglía/patología , Microglía/efectos de la radiación , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mutación Missense , Proteolisis/efectos de la radiación
10.
J Biomol Struct Dyn ; 38(3): 807-826, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-30836878

RESUMEN

Aryl Hydrocarbon Receptor (AhR) is a key player to regulate the expression of a group of enzymes known as cytochrome P450s (CYPs) super family (CYP1A1, CYP1B1, CYP2B6, and CYP2E1) which metabolites diverse endogenous as well as toxic compounds such as Benzo[a] Pyrene (B[a] P) and TCDD. B[a] P induces oxidative stress and causes degeneration of dopaminergic neurons in the midbrain, may leads to Parkinson's disease (PD). The metabolism of B[a] P through the expression of CYPs is mainly triggered after binding of B[a] P within ligand binding domain of AhR. But, the molecular mechanism of AhR mediated xenobiotic metabolism in presence of diverse phytochemicals is yet to be studied. The solved AhR (PDB ID: 5NJ8, 23-273aa) structure lacks information for ligand binding domain therefore both wild type and mutant models were predicted and screened virtually against sixty one natural compounds. The result proposed withaferin A, withanolide A, withanolide B, withanolide D and withanone of plant Withania Somnifera as efficient ligand against both wild type and mutants (V381A and V381D) AhR models. However, in silico studies hypothesised withanolide A as a potent phytochemical to trigger the AhR mediated gene regulation activity of CYPs. The in vivo study in zebra fish model proposed about the neuro protective role of W. Somnifera leaf extract in presence of B[a]P. The present study would throw lights on the molecular mechanism of phytochemicals mediated AhR activity which may be useful in treatment of PD. [Formula: see text] Communicated by Ramaswamy H. Sarma.


Asunto(s)
Simulación por Computador , Proteínas Mutantes/metabolismo , Enfermedad de Parkinson/tratamiento farmacológico , Fitoquímicos/uso terapéutico , Receptores de Hidrocarburo de Aril/metabolismo , Secuencia de Aminoácidos , Animales , Encéfalo/efectos de los fármacos , Encéfalo/enzimología , Citocromo P-450 CYP1A1/metabolismo , Evaluación Preclínica de Medicamentos , Humanos , Enlace de Hidrógeno , Ligandos , Simulación del Acoplamiento Molecular , Proteínas Mutantes/química , Fitoquímicos/química , Fitoquímicos/farmacología , Unión Proteica , Dominios Proteicos , Mapas de Interacción de Proteínas , Estabilidad Proteica , Estructura Secundaria de Proteína , Receptores de Hidrocarburo de Aril/química , Pez Cebra
11.
Nature ; 575(7781): 203-209, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31666698

RESUMEN

Accumulation of mutant proteins is a major cause of many diseases (collectively called proteopathies), and lowering the level of these proteins can be useful for treatment of these diseases. We hypothesized that compounds that interact with both the autophagosome protein microtubule-associated protein 1A/1B light chain 3 (LC3)1 and the disease-causing protein may target the latter for autophagic clearance. Mutant huntingtin protein (mHTT) contains an expanded polyglutamine (polyQ) tract and causes Huntington's disease, an incurable neurodegenerative disorder2. Here, using small-molecule-microarray-based screening, we identified four compounds that interact with both LC3 and mHTT, but not with the wild-type HTT protein. Some of these compounds targeted mHTT to autophagosomes, reduced mHTT levels in an allele-selective manner, and rescued disease-relevant phenotypes in cells and in vivo in fly and mouse models of Huntington's disease. We further show that these compounds interact with the expanded polyQ stretch and could lower the level of mutant ataxin-3 (ATXN3), another disease-causing protein with an expanded polyQ tract3. This study presents candidate compounds for lowering mHTT and potentially other disease-causing proteins with polyQ expansions, demonstrating the concept of lowering levels of disease-causing proteins using autophagosome-tethering compounds.


Asunto(s)
Alelos , Evaluación Preclínica de Medicamentos/métodos , Proteína Huntingtina/antagonistas & inhibidores , Proteína Huntingtina/genética , Proteínas Mutantes/antagonistas & inhibidores , Proteínas Mutantes/genética , Mutación/genética , Animales , Ataxina-3/genética , Autofagosomas/metabolismo , Autofagia , Modelos Animales de Enfermedad , Proteínas de Drosophila/antagonistas & inhibidores , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Femenino , Humanos , Proteína Huntingtina/química , Proteína Huntingtina/metabolismo , Masculino , Ratones , Proteínas Asociadas a Microtúbulos/genética , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Mutación/efectos de los fármacos , Neuronas/citología , Péptidos/genética , Fenotipo , Reproducibilidad de los Resultados
12.
Sci Rep ; 9(1): 6915, 2019 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-31061434

RESUMEN

Limb-girdle muscular dystrophy type 2D (LGMD2D) is characterized by a progressive proximal muscle weakness. LGMD2D is caused by mutations in the gene encoding α-sarcoglycan (α-SG), a dystrophin-associated glycoprotein that plays a key role in the maintenance of sarcolemma integrity in striated muscles. We report here on the development of a new in vitro high-throughput screening assay that allows the monitoring of the proper localization of the most prevalent mutant form of α-SG (R77C substitution). Using this assay, we screened a library of 2560 FDA-approved drugs and bioactive compounds and identified thiostrepton, a cyclic antibiotic, as a potential drug to repurpose for LGMD2D treatment. Characterization of the thiostrepton effect revealed a positive impact on R77C-α-SG and other missense mutant protein localization (R34H, I124T, V247M) in fibroblasts overexpressing these proteins. Finally, further investigations of the molecular mechanisms of action of the compound revealed an inhibition of the chymotrypsin-like activity of the proteasome 24 h after thiostrepton treatment and a synergistic effect with bortezomib, an FDA-approved proteasome inhibitor. This study reports on the first in vitro model for LGMD2D that is compatible with high-throughput screening and proposes a new therapeutic option for LGMD2D caused by missense mutations of α-SG.


Asunto(s)
Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Pliegue de Proteína/efectos de los fármacos , Proteolisis/efectos de los fármacos , Sarcoglicanos/química , Sarcoglicanos/metabolismo , Tioestreptona/farmacología , Línea Celular , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Evaluación Preclínica de Medicamentos , Humanos , Células Madre Pluripotentes Inducidas/citología , Proteínas Mutantes/genética , Mioblastos/citología , Mioblastos/efectos de los fármacos , Sarcoglicanos/genética
13.
J Natl Cancer Inst ; 111(11): 1202-1215, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30990221

RESUMEN

BACKGROUND: Anti-tumorigenic vs pro-tumorigenic roles of estrogen receptor-beta (ESR2) in breast cancer remain unsettled. We investigated the potential of TP53 status to be a determinant of the bi-faceted role of ESR2 and associated therapeutic implications for triple negative breast cancer (TNBC). METHODS: ESR2-TP53 interaction was analyzed with multiple assays including the in situ proximity ligation assay. Transcriptional effects on TP53-target genes and cell proliferation in response to knocking down or overexpressing ESR2 were determined. Patient survival according to ESR2 expression levels and TP53 mutation status was analyzed in the basal-like TNBC subgroup in the Molecular Taxonomy of Breast Cancer International Consortium (n = 308) and Roswell Park Comprehensive Cancer Center (n = 46) patient cohorts by univariate Cox regression and log-rank test. All statistical tests are two-sided. RESULTS: ESR2 interaction with wild-type and mutant TP53 caused pro-proliferative and anti-proliferative effects, respectively. Depleting ESR2 in cells expressing wild-type TP53 resulted in increased expression of TP53-target genes CDKN1A (control group mean [SD] = 1 [0.13] vs ESR2 depletion group mean [SD] = 2.08 [0.24], P = .003) and BBC3 (control group mean [SD] = 1 [0.06] vs ESR2 depleted group mean [SD] = 1.92 [0.25], P = .003); however, expression of CDKN1A (control group mean [SD] = 1 [0.21] vs ESR2 depleted group mean [SD] = 0.56 [0.12], P = .02) and BBC3 (control group mean [SD] = 1 [0.03] vs ESR2 depleted group mean [SD] = 0.55 [0.09], P = .008) was decreased in cells expressing mutant TP53. Overexpressing ESR2 had opposite effects. Tamoxifen increased ESR2-mutant TP53 interaction, leading to reactivation of TP73 and apoptosis. High levels of ESR2 expression in mutant TP53-expressing basal-like tumors is associated with better prognosis (Molecular Taxonomy of Breast Cancer International Consortium cohort: log-rank P = .001; hazard ratio = 0.26, 95% confidence interval = 0.08 to 0.84, univariate Cox P = .02). CONCLUSIONS: TP53 status is a determinant of the functional duality of ESR2. Our study suggests that ESR2-mutant TP53 combination prognosticates survival in TNBC revealing a novel strategy to stratify TNBC for therapeutic intervention potentially by repurposing tamoxifen.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Carcinogénesis/patología , Receptor beta de Estrógeno/metabolismo , Proteínas Mutantes/metabolismo , Mutación , Neoplasias de la Mama Triple Negativas/patología , Proteína p53 Supresora de Tumor/metabolismo , Biomarcadores de Tumor/genética , Carcinogénesis/genética , Carcinogénesis/metabolismo , Proliferación Celular , Estudios de Cohortes , Receptor beta de Estrógeno/genética , Femenino , Humanos , Proteínas Mutantes/genética , Pronóstico , Tasa de Supervivencia , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/metabolismo , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/genética
14.
Clin Exp Med ; 19(2): 201-210, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30523507

RESUMEN

The purpose of the present study was to investigate the in vitro and in vivo activity of PLX9486, a tyrosine kinase inhibitor (TKI) targeting both primary KIT exon 9 and 11 and secondary exon 17 and 18 mutations in gastrointestinal stromal tumors (GISTs). Imatinib, a potent inhibitor of mutated KIT, has revolutionized the clinical management of advanced, metastatic GIST. However, secondary resistance develops mainly through acquired mutations in KIT exons 13/14 or exons 17/18. Second-line sunitinib potently inhibits KIT exon 13/14 mutants but is ineffective against exon 17 mutations. In our study, PLX9486 demonstrated in vitro nanomolar potency in inhibiting the growth and KIT phosphorylation of engineered BaF3 cells transformed with KIT exon 17 mutations (p.D816V) and with the double KIT exon 11/17 mutations (p.V560G/D816V). The in vivo efficacy of PLX9486 was evaluated using two imatinib-resistant GIST patient-derived xenograft (PDX) models. In UZLX-GIST9 (KIT: p.P577del;W557LfsX5;D820G), PLX9486 100 mg/kg/day resulted in significant inhibition of proliferation. Pharmacodynamic analysis showed a pronounced reduction in mitogen-activated protein kinase (MAPK) activation and other downstream effects of the KIT signaling pathway but no significant effect on KIT Y703 and Y719 phosphorylation. Similarly, in MRL-GIST1 (KIT: p.W557_K558del;Y823D) PLX9486 treatment led to significant tumor regression and strong inhibition of MAPK activation. Interestingly, the inhibitory effect on MAPK activation was evident even after a single dose of PLX9486. In conclusion, PLX9486 showed anti-tumor efficacy in patient-derived imatinib-resistant GIST xenograft models, mainly through inhibition of KIT signaling. These preclinical efficacy data encourage further testing of PLX9486 in the clinical setting.


Asunto(s)
Antineoplásicos/administración & dosificación , Inhibidores Enzimáticos/administración & dosificación , Tumores del Estroma Gastrointestinal/tratamiento farmacológico , Tumores del Estroma Gastrointestinal/patología , Proteínas Mutantes/genética , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-kit/genética , Animales , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Xenoinjertos , Humanos , Ratones , Proteínas Mutantes/metabolismo , Trasplante de Neoplasias , Proteínas Proto-Oncogénicas c-kit/metabolismo , Resultado del Tratamiento
15.
Elife ; 72018 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-30136928

RESUMEN

Amyotrophic lateral sclerosis (ALS) is characterized by the progressive degeneration of motoneurons in the primary motor cortex (pMO) and in spinal cord. However, the pathogenic process involves multiple subnetworks in the brain and functional MRI studies demonstrate an increase in functional connectivity in areas connected to pMO despite the ongoing neurodegeneration. The extent and the structural basis of the motor subnetwork remodeling in experimentally tractable models remain unclear. We have developed a new retrograde AAV9 to quantitatively map the projections to pMO in the SOD1(G93A) ALS mouse model. We show an increase in the number of neurons projecting from somatosensory cortex to pMO at presymptomatic stages, followed by an increase in projections from thalamus, auditory cortex and contralateral MO (inputs from 20 other structures remains unchanged) as disease advances. The stage- and structure-dependent remodeling of projection to pMO in ALS may provide insights into the hyperconnectivity observed in ALS patients.


Asunto(s)
Esclerosis Amiotrófica Lateral/fisiopatología , Dependovirus/metabolismo , Corteza Motora/fisiopatología , Esclerosis Amiotrófica Lateral/patología , Animales , Espinas Dendríticas/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Humanos , Masculino , Ratones , Corteza Motora/patología , Proteínas Mutantes/metabolismo , Red Nerviosa/patología , Red Nerviosa/fisiopatología , Pliegue de Proteína , Células Piramidales/metabolismo , Células Piramidales/patología , Superóxido Dismutasa/metabolismo , Tálamo/patología , Tálamo/fisiopatología
16.
Am J Hum Genet ; 102(6): 1126-1142, 2018 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-29805043

RESUMEN

The proteasome processes proteins to facilitate immune recognition and host defense. When inherently defective, it can lead to aberrant immunity resulting in a dysregulated response that can cause autoimmunity and/or autoinflammation. Biallelic or digenic loss-of-function variants in some of the proteasome subunits have been described as causing a primary immunodeficiency disease that manifests as a severe dysregulatory syndrome: chronic atypical neutrophilic dermatosis with lipodystrophy and elevated temperature (CANDLE). Proteasome maturation protein (POMP) is a chaperone for proteasome assembly and is critical for the incorporation of catalytic subunits into the proteasome. Here, we characterize and describe POMP-related autoinflammation and immune dysregulation disease (PRAID) discovered in two unrelated individuals with a unique constellation of early-onset combined immunodeficiency, inflammatory neutrophilic dermatosis, and autoimmunity. We also begin to delineate a complex genetic mechanism whereby de novo heterozygous frameshift variants in the penultimate exon of POMP escape nonsense-mediated mRNA decay (NMD) and result in a truncated protein that perturbs proteasome assembly by a dominant-negative mechanism. To our knowledge, this mechanism has not been reported in any primary immunodeficiencies, autoinflammatory syndromes, or autoimmune diseases. Here, we define a unique hypo- and hyper-immune phenotype and report an immune dysregulation syndrome caused by frameshift mutations that escape NMD.


Asunto(s)
Predisposición Genética a la Enfermedad , Chaperonas Moleculares/genética , Mutación/genética , Degradación de ARNm Mediada por Codón sin Sentido/genética , Secuencia de Bases , Línea Celular , Estrés del Retículo Endoplásmico , Exones/genética , Familia , Mutación del Sistema de Lectura/genética , Heterocigoto , Humanos , Síndromes de Inmunodeficiencia/genética , Inmunofenotipificación , Recién Nacido , Inflamación/patología , Interferón Tipo I/metabolismo , Masculino , Proteínas Mutantes/metabolismo , Fenotipo , Complejo de la Endopetidasa Proteasomal/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Síndrome , Respuesta de Proteína Desplegada
17.
Mol Genet Metab ; 123(4): 449-462, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29526616

RESUMEN

Oxidative stress is a known contributing factor in mitochondrial respiratory chain (RC) disease pathogenesis. Yet, no efficient means exists to objectively evaluate the comparative therapeutic efficacy or toxicity of different antioxidant compounds empirically used in human RC disease. We postulated that pre-clinical comparative analysis of diverse antioxidant drugs having suggested utility in primary RC disease using animal and cellular models of RC dysfunction may improve understanding of their integrated effects and physiologic mechanisms, and enable prioritization of lead antioxidant molecules to pursue in human clinical trials. Here, lifespan effects of N-acetylcysteine (NAC), vitamin E, vitamin C, coenzyme Q10 (CoQ10), mitochondrial-targeted CoQ10 (MS010), lipoate, and orotate were evaluated as the primary outcome in a well-established, short-lived C. elegans gas-1(fc21) animal model of RC complex I disease. Healthspan effects were interrogated to assess potential reversal of their globally disrupted in vivo mitochondrial physiology, transcriptome profiles, and intermediary metabolic flux. NAC or vitamin E fully rescued, and coenzyme Q, lipoic acid, orotic acid, and vitamin C partially rescued gas-1(fc21) lifespan toward that of wild-type N2 Bristol worms. MS010 and CoQ10 largely reversed biochemical pathway expression changes in gas-1(fc21) worms. While nearly all drugs normalized the upregulated expression of the "cellular antioxidant pathway", they failed to rescue the mutant worms' increased in vivo mitochondrial oxidant burden. NAC and vitamin E therapeutic efficacy were validated in human fibroblast and/or zebrafish complex I disease models. Remarkably, rotenone-induced zebrafish brain death was preventable partially with NAC and fully with vitamin E. Overall, these pre-clinical model animal data demonstrate that several classical antioxidant drugs do yield significant benefit on viability and survival in primary mitochondrial disease, where their major therapeutic benefit appears to result from targeting global cellular, rather than intramitochondria-specific, oxidative stress. Clinical trials are needed to evaluate whether the two antioxidants, NAC and vitamin E, that show greatest efficacy in translational model animals significantly improve the survival, function, and feeling of human subjects with primary mitochondrial RC disease.


Asunto(s)
Acetilcisteína/farmacología , Evaluación Preclínica de Medicamentos , Complejo I de Transporte de Electrón/metabolismo , Longevidad , Enfermedades Mitocondriales/tratamiento farmacológico , Estrés Oxidativo/efectos de los fármacos , Vitamina E/farmacología , Animales , Animales Modificados Genéticamente , Antioxidantes/farmacología , Caenorhabditis elegans , Células Cultivadas , Complejo I de Transporte de Electrón/genética , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/patología , Depuradores de Radicales Libres/farmacología , Humanos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mitocondrias/patología , Enfermedades Mitocondriales/genética , Enfermedades Mitocondriales/metabolismo , Enfermedades Mitocondriales/patología , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mutación
18.
FASEB J ; 32(1): 404-416, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28928244

RESUMEN

Zinc, an essential micronutrient, has a cancer preventive role. Zinc deficiency has been shown to contribute to the progression of esophageal cancer. Orai1, a store-operated Ca2+ entry (SOCE) channel, was previously reported to be highly expressed in tumor tissues removed from patients with esophageal squamous cell carcinoma (ESCC) with poor prognosis, and elevation of its expression contributes to both hyperactive intracellular Ca2+ oscillations and fast cell proliferation in human ESCC cells. However, the molecular basis of cancer preventive functions of zinc and its association with Orai1-mediated cell proliferation remains unknown. The present study shows that zinc supplementation significantly inhibits proliferation of ESCC cell lines and that the effect of zinc is reversible with N,N,N',N'-tetrakis (2-pyridylmethyl) ethylenediamine, a specific Zn2+ chelator, whereas nontumorigenic esophageal epithelial cells are significantly less sensitive to zinc treatment. Fluorescence live cell imaging revealed that extracellular Zn2+ exerted rapid inhibitory effects on Orai1-mediated SOCE and on intracellular Ca2+ oscillations in the ESCC cells. Knockdown of Orai1 or expression of Orai1 mutants with compromised zinc binding significantly diminished sensitivity of the cancer cells to zinc treatment in both SOCE and cell proliferation analyses. These data suggest that zinc may inhibit cell proliferation of esophageal cancer cells through Orai1-mediated intracellular Ca2+ oscillations and reveal a possible molecular basis for zinc-induced cancer prevention and Orai1-SOCE signaling pathway in cancer cells.-Choi, S., Cui, C., Luo, Y., Kim, S.-H., Ko, J.-K., Huo, X., Ma, J., Fu, L.-W., Souza, R. F., Korichneva, I., Pan, Z. Selective inhibitory effects of zinc on cell proliferation in esophageal squamous cell carcinoma through Orai1.


Asunto(s)
Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/metabolismo , Neoplasias Esofágicas/tratamiento farmacológico , Neoplasias Esofágicas/metabolismo , Proteína ORAI1/metabolismo , Zinc/farmacología , Sustitución de Aminoácidos , Señalización del Calcio/efectos de los fármacos , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Quelantes/farmacología , Neoplasias Esofágicas/patología , Carcinoma de Células Escamosas de Esófago , Etilenodiaminas/farmacología , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Humanos , Modelos Biológicos , Mutagénesis Sitio-Dirigida , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Proteína ORAI1/antagonistas & inhibidores , Proteína ORAI1/genética
19.
Elife ; 62017 10 24.
Artículo en Inglés | MEDLINE | ID: mdl-29063836

RESUMEN

Mutations in the human BEST1 gene lead to retinal degenerative diseases displaying progressive vision loss and even blindness. BESTROPHIN1, encoded by BEST1, is predominantly expressed in retinal pigment epithelium (RPE), but its physiological role has been a mystery for the last two decades. Using a patient-specific iPSC-based disease model and interdisciplinary approaches, we comprehensively analyzed two distinct BEST1 patient mutations, and discovered mechanistic correlations between patient clinical phenotypes, electrophysiology in their RPEs, and the structure and function of BESTROPHIN1 mutant channels. Our results revealed that the disease-causing mechanism of BEST1 mutations is centered on the indispensable role of BESTROPHIN1 in mediating the long speculated Ca2+-dependent Cl- current in RPE, and demonstrate that the pathological potential of BEST1 mutations can be evaluated and predicted with our iPSC-based 'disease-in-a-dish' approach. Moreover, we demonstrated that patient RPE is rescuable with viral gene supplementation, providing a proof-of-concept for curing BEST1-associated diseases.


Asunto(s)
Bestrofinas/genética , Bestrofinas/metabolismo , Calcio/metabolismo , Cloruros/metabolismo , Mutación Missense , Enfermedades de la Retina/fisiopatología , Epitelio Pigmentado de la Retina/fisiología , Anciano , Bestrofinas/química , Células Cultivadas , Niño , Cristalografía por Rayos X , Humanos , Iones/metabolismo , Masculino , Modelos Moleculares , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Conformación Proteica , Enfermedades de la Retina/genética
20.
Nat Chem Biol ; 13(11): 1179-1186, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28920930

RESUMEN

The GATOR1 (SEACIT) complex consisting of Iml1-Npr2-Npr3 inhibits target of rapamycin complex 1 (TORC1) in response to amino acid insufficiency. In glucose medium, Saccharomyces cerevisiae mutants lacking the function of this complex grow poorly in the absence of amino acid supplementation, despite showing hallmarks of increased TORC1 signaling. Such mutants sense that they are amino acid replete and thus repress metabolic activities that are important for achieving this state. We found that npr2Δ mutants have defective mitochondrial tricarboxylic acid (TCA)-cycle activity and retrograde response. Supplementation with glutamine, and especially aspartate, which are nitrogen-containing forms of TCA-cycle intermediates, rescues growth of npr2Δ mutants. These amino acids are then consumed in biosynthetic pathways that require nitrogen to support proliferative metabolism. Our findings revealed that negative regulators of TORC1, such as GATOR1 (SEACIT), regulate the cataplerotic synthesis of these amino acids from the TCA cycle, in tune with the amino acid and nitrogen status of cells.


Asunto(s)
Ciclo del Ácido Cítrico , Mitocondrias/metabolismo , Complejos Multiproteicos/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Ácido Aspártico/metabolismo , Glutamina/metabolismo , Complejos Multiproteicos/genética , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Nitrógeno/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/crecimiento & desarrollo , Proteínas de Saccharomyces cerevisiae/genética , Transducción de Señal , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA