Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 341
Filtrar
1.
Int J Mol Sci ; 22(16)2021 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-34445333

RESUMEN

Lynch syndrome (LS) is one of the most common hereditary cancer predisposition syndromes worldwide. Individuals with LS have a high risk of developing colorectal or endometrial cancer, as well as several other cancers. LS is caused by autosomal dominant pathogenic variants in one of the DNA mismatch repair (MMR) genes MLH1, MSH2, PMS2 or MSH6, and typically include truncating variants, such as frameshift, nonsense or splicing variants. However, a significant number of missense, intronic, or silent variants, or small in-frame insertions/deletions, are detected during genetic screening of the MMR genes. The clinical effects of these variants are often more difficult to predict, and a large fraction of these variants are classified as variants of uncertain significance (VUS). It is pivotal for the clinical management of LS patients to have a clear genetic diagnosis, since patients benefit widely from screening, preventive and personal therapeutic measures. Moreover, in families where a pathogenic variant is identified, testing can be offered to family members, where non-carriers can be spared frequent surveillance, while carriers can be included in cancer surveillance programs. It is therefore important to reclassify VUSs, and, in this regard, functional assays can provide insight into the effect of a variant on the protein or mRNA level. Here, we briefly describe the disorders that are related to MMR deficiency, as well as the structure and function of MSH6. Moreover, we review the functional assays that are used to examine VUS identified in MSH6 and discuss the results obtained in relation to the ACMG/AMP PS3/BS3 criterion. We also provide a compiled list of the MSH6 variants examined by these assays. Finally, we provide a future perspective on high-throughput functional analyses with specific emphasis on the MMR genes.


Asunto(s)
Proteínas de Unión al ADN/genética , Técnicas Genéticas , Animales , Proteínas de Unión al ADN/clasificación , Proteínas de Unión al ADN/fisiología , Pruebas Genéticas/métodos , Humanos , Proteínas Mutantes/clasificación , Proteínas Mutantes/genética , Proteínas Mutantes/fisiología , Isoformas de Proteínas/clasificación , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiología , Empalme del ARN/genética
2.
J Virol ; 95(21): e0131021, 2021 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-34379504

RESUMEN

Dengue virus (DENV) constitutes one of the most important arboviral pathogens affecting humans. The high prevalence of DENV infections, which cause more than 20,000 deaths annually, and the lack of effective vaccines or direct-acting antiviral drugs make it a global health concern. DENV genome replication occurs in close association with the host endomembrane system, which is remodeled to form the viral replication organelle that originates from endoplasmic reticulum (ER) membranes. To date, the viral and cellular determinants responsible for the biogenesis of DENV replication organelles are still poorly defined. The viral nonstructural protein 4A (NS4A) can remodel membranes and has been shown to associate with numerous host factors in DENV-replicating cells. In the present study, we used reverse and forward genetic screens and identified sites within NS4A required for DENV replication. We also mapped the determinants in NS4A required for interactions with other viral proteins. Moreover, taking advantage of our recently developed polyprotein expression system, we evaluated the role of NS4A in the formation of DENV replication organelles. Together, we report a detailed map of determinants within NS4A required for RNA replication, interaction with other viral proteins, and replication organelle formation. Our results suggest that NS4A might be an attractive target for antiviral therapy. IMPORTANCE DENV is the most prevalent mosquito-borne virus, causing around 390 million infections each year. There are no approved therapies to treat DENV infection, and the only available vaccine shows limited efficacy. The viral nonstructural proteins have emerged as attractive drug targets due to their pivotal role in RNA replication and establishment of virus-induced membranous compartments, designated replication organelles (ROs). The transmembrane protein NS4A, generated by cleavage of the NS4A-2K-4B precursor, contributes to DENV replication by unknown mechanisms. Here, we report a detailed genetic interaction map of NS4A and identify residues required for RNA replication and interaction between NS4A-2K-4B and NS2B-3 as well as NS1. Importantly, by means of an expression-based system, we demonstrate the essential role of NS4A in RO biogenesis and identify determinants in NS4A required for this process. Our data suggest that NS4A is an attractive target for antiviral therapy.


Asunto(s)
Virus del Dengue/fisiología , Dengue/virología , Biogénesis de Organelos , Orgánulos/virología , Proteínas no Estructurales Virales/fisiología , Secuencia de Aminoácidos , Animales , Línea Celular , Chlorocebus aethiops , Virus del Dengue/ultraestructura , Interacciones Microbiota-Huesped , Humanos , Proteínas Mutantes/fisiología , Mutación , Orgánulos/ultraestructura , Unión Proteica , ARN/metabolismo , ARN Viral , Genética Inversa/métodos , Células Vero , Replicación Viral
3.
Acta Biochim Biophys Sin (Shanghai) ; 53(4): 463-471, 2021 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-33751023

RESUMEN

A large number of proteins involved in RNA metabolism possess a double-stranded RNA-binding domain (dsRBD), whose sequence variations and functional versatilities are still being recognized. All dsRBDs have a similar structural fold: α1-L1-ß1-L2-ß2-L3-ß3-L4-α2 (α represents an α-helix, ß a ß-sheet, and L a loop conformation between the well-defined secondary structures). Our recent work revealed that the dsRBD in Drosha, which is involved in animal microRNA (miRNA) biogenesis, differs from other dsRBDs by containing a short insertion in its L1 region and that this insertion is important for Drosha function. We asked why the same insertion is excluded in all other dsRBDs and proposed that a longer L1 may be detrimental to their functions. In this study, to test this hypothesis, we inserted the Drosha sequence into several well-known dsRBDs from various organisms. Gel mobility shift assay demonstrated that L1 extension invariably reduced RNA binding by these dsRBDs. In addition, such a mutation in Dicer, another protein involved in miRNA biogenesis, impaired Dicer's ability to process miRNAs, which led to de-repression of reporter expression, in human cells. Taken together, our results add to the growing appreciation of the diversity in dsRBDs and suggest that dsRBDs have intricate structures and functions that are sensitive to perturbations in the L1 region.


Asunto(s)
Motivo de Unión al ARN Bicatenario , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/fisiología , Secuencia de Aminoácidos , Animales , ARN Helicasas DEAD-box/química , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/fisiología , ADN de Cadena Simple/metabolismo , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Proteínas de Drosophila/fisiología , Células HEK293 , Humanos , MicroARNs/metabolismo , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/fisiología , Estructura Secundaria de Proteína , ARN/metabolismo , Proteínas de Unión al ARN/genética , Ribonucleasa III/química , Ribonucleasa III/genética , Ribonucleasa III/fisiología , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/fisiología , Proteínas de Xenopus/química , Proteínas de Xenopus/genética , Proteínas de Xenopus/fisiología
4.
Cancer Res ; 81(4): 935-944, 2021 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-33323382

RESUMEN

p53 is a short-lived protein with low basal levels under normal homeostasis conditions. However, upon DNA damage, levels of p53 dramatically increase for its activation. Although robust stabilization of p53 serves as a "trademark" for DNA damage responses, the requirement for such dramatic protein stabilization in tumor suppression has not been well addressed. Here we generated a mutant p53KQ mouse where all the C-terminal domain lysine residues were mutated to glutamines (K to Q mutations at K367, K369, K370, K378, K379, K383, and K384) to mimic constitutive acetylation of the p53 C-terminus. Because of p53 activation, p53KQ/KQ mice were perinatal lethal, yet this lethality was averted in p53KQ/- mice, which displayed normal postnatal development. Nevertheless, p53KQ/- mice died prematurely due to anemia and hematopoiesis failure. Further analyses indicated that expression of the acetylation-mimicking p53 mutant in vivo induces activation of p53 targets in various tissues without obviously increasing p53 levels. In the well-established pancreatic ductal adenocarcinoma (PDAC) mouse model, expression of the acetylation-mimicking p53-mutant protein effectively suppressed K-Ras-induced PDAC development in the absence of robust p53 stabilization. Together, our results provide proof-of-principle evidence that p53-mediated transcriptional function and tumor suppression can be achieved independently of its robust stabilization and reveal an alternative approach to activate p53 function for therapeutic purposes. SIGNIFICANCE: Although robust p53 stabilization is critical for acute p53 responses such as DNA damage, this study underscores the important role of low basal p53 protein levels in p53 activation and tumor suppression.


Asunto(s)
Proteína p53 Supresora de Tumor/metabolismo , Proteína p53 Supresora de Tumor/fisiología , Acetilación , Animales , Apoptosis/genética , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patología , Células Cultivadas , Daño del ADN/genética , Genes Supresores de Tumor/fisiología , Lisina/metabolismo , Ratones , Ratones Transgénicos , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Proteínas Mutantes/fisiología , Mutación , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Procesamiento Proteico-Postraduccional/genética , Estabilidad Proteica , Activación Transcripcional/genética , Proteína p53 Supresora de Tumor/química
5.
Endocrinology ; 162(3)2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33300995

RESUMEN

Androgen receptor (AR) signaling continues to drive castration-resistant prostate cancer (CRPC) in spite of androgen deprivation therapy (ADT). Constitutively active shorter variants of AR, lacking the ligand binding domain, are frequently expressed in CRPC and have emerged as a potential mechanism for prostate cancer to escape ADT. ARv7 and ARv567es are 2 of the most commonly detected variants of AR in clinical samples of advanced, metastatic prostate cancer. It is not clear if variants of AR merely act as weaker substitutes for AR or can mediate unique isoform-specific activities different from AR. In this study, we employed LNCaP prostate cancer cell lines with inducible expression of ARv7 or ARv567es to delineate similarities and differences in transcriptomics, metabolomics, and lipidomics resulting from the activation of AR, ARv7, or ARv567es. While the majority of target genes were similarly regulated by the action of all 3 isoforms, we found a clear difference in transcriptomic activities of AR versus the variants, and a few differences between ARv7 and ARv567es. Some of the target gene regulation by AR isoforms was similar in the VCaP background as well. Differences in downstream activities of AR isoforms were also evident from comparison of the metabolome and lipidome in an LNCaP model. Overall our study implies that shorter variants of AR are capable of mediating unique downstream activities different from AR and some of these are isoform specific.


Asunto(s)
Neoplasias de la Próstata Resistentes a la Castración/genética , Receptores Androgénicos/fisiología , Empalme Alternativo/genética , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica/genética , Células HEK293 , Humanos , Metabolismo de los Lípidos/genética , Masculino , Proteínas Mutantes/fisiología , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/patología , Isoformas de Proteínas/fisiología , Receptores Androgénicos/química , Receptores Androgénicos/genética
6.
Biomolecules ; 10(11)2020 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-33238579

RESUMEN

The bacterial RNA polymerase (RNAP) is a multi-subunit protein complex (α2ßß'ω σ) containing the smallest subunit, ω. Although identified early in RNAP research, its function remained ambiguous and shrouded with controversy for a considerable period. It was shown before that the protein has a structural role in maintaining the conformation of the largest subunit, ß', and its recruitment in the enzyme assembly. Despite evolutionary conservation of ω and its role in the assembly of RNAP, E. coli mutants lacking rpoZ (codes for ω) are viable due to the association of the global chaperone protein GroEL with RNAP. To get a better insight into the structure and functional role of ω during transcription, several dominant lethal mutants of ω were isolated. The mutants showed higher binding affinity compared to that of native ω to the α2ßß' subassembly. We observed that the interaction between α2ßß' and these lethal mutants is driven by mostly favorable enthalpy and a small but unfavorable negative entropy term. However, during the isolation of these mutants we isolated a silent mutant serendipitously, which showed a lethal phenotype. Silent mutant of a given protein is defined as a protein having the same sequence of amino acids as that of wild type but having mutation in the gene with alteration in base sequence from more frequent code to less frequent one due to codon degeneracy. Eventually, many silent mutants were generated to understand the role of rare codons at various positions in rpoZ. We observed that the dominant lethal mutants of ω having either point mutation or silent in nature are more structured in comparison to the native ω. However, the silent code's position in the reading frame of rpoZ plays a role in the structural alteration of the translated protein. This structural alteration in ω makes it more rigid, which affects the plasticity of the interacting domain formed by ω and α2ßß'. Here, we attempted to describe how the conformational flexibility of the ω helps in maintaining the plasticity of the active site of RNA polymerase. The dominant lethal mutant of ω has a suppressor mapped near the catalytic center of the ß' subunit, and it is the same for both types of mutants.


Asunto(s)
ARN Polimerasas Dirigidas por ADN/química , ARN Polimerasas Dirigidas por ADN/fisiología , Proteínas Bacterianas/química , Proteínas Bacterianas/fisiología , Proteínas Mutantes/química , Proteínas Mutantes/fisiología , Subunidades de Proteína/química , Subunidades de Proteína/fisiología , Relación Estructura-Actividad , Factores de Transcripción/metabolismo
7.
Biochimie ; 177: 87-97, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32828823

RESUMEN

Emerging observations suggest that ribosomal proteins (RPs) play important extra-ribosomal roles in maintenance of cellular homeostasis. However, the mechanistic insights into these processes have not been extensively explored, especially in pathogenic bacteria. Here, we present our findings on potential extra-ribosomal functions of Mycobacterium tuberculosis (Mtb) RPs. We observed that Mtb RpsB and RpsQ are differentially localized to cell wall fraction in M. tuberculosis (H37Rv), while their M. smegmatis (Msm) homologs are primarily cytosolic. Cellular fractionation of ectopically expressed Mtb RPs in surrogate host (M. smegmatis) also shows their association with cell membrane/cell wall without any gross changes in cell morphology. M. smegmatis expressing Mtb RpsB exhibited altered redox homeostasis, decreased drug-induced ROS, reduced cell wall permeability and increased tolerance to various proteotoxic stress (oxidative stress, SDS and starvation). Mtb RpsB expression was also associated with increased resistance specifically towards Isoniazid, Ethionamide and Streptomycin. The enhanced drug tolerance was specific to Mtb RpsB and not observed upon ectopic expression of M. smegmatis homolog (Msm RpsB). Interestingly, C-terminus deletion in Mtb RpsB affected its localization and reversed the stress-resilient phenotypes. We also observed that M. tuberculosis (H37Rv) with upregulated RpsB levels had higher intracellular survival in macrophage. All these observations hint towards existence of moonlighting roles of Mtb RpsB in imparting stress resilience to mycobacteria. This work open avenues for further exploration of alternative pathways associated with fitness and drug tolerance in mycobacteria.


Asunto(s)
Proteínas Bacterianas/fisiología , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/metabolismo , Proteínas Ribosómicas/fisiología , Antibacterianos/farmacología , Proteínas Bacterianas/química , Membrana Celular/metabolismo , Pared Celular/metabolismo , Citosol/metabolismo , Tolerancia a Medicamentos/genética , Humanos , Lípidos/análisis , Macrófagos/metabolismo , Macrófagos/microbiología , Proteínas Mutantes/química , Proteínas Mutantes/fisiología , Mycobacterium smegmatis/efectos de los fármacos , Mycobacterium smegmatis/crecimiento & desarrollo , Mycobacterium smegmatis/metabolismo , Oxidación-Reducción , Estrés Oxidativo/genética , Permeabilidad , Especies Reactivas de Oxígeno/metabolismo , Proteínas Ribosómicas/química , Ribosomas/química , Células THP-1
8.
PLoS One ; 15(6): e0234394, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32574176

RESUMEN

In the BACHD mouse model of Huntington's disease (HD), deletion of the N17 domain of the Huntingtin gene (BACHDΔN17, Q97) has been reported to lead to nuclear accumulation of mHTT and exacerbation of motor deficits, neuroinflammation and striatal atrophy (Gu et al., 2015). Here we characterized the effect of N17 deletion on dorsolateral striatal medium spiny neurons (MSNs) in BACHDΔN17 (Q97) and BACWTΔN17 (Q31) mice by comparing them to MSNs in wildtype (WT) mice. Mice were characterized on a series of motor tasks and subsequently whole cell patch clamp recordings with simultaneous biocytin filling of MSNs in in vitro striatal slices from these mice were used to comprehensively assess their physiological and morphological features. Key findings include that: Q97 mice exhibit impaired gait and righting reflexes but normal tail suspension reflexes and normal coats while Q31 mice do not differ from WT; intrinsic membrane and action potential properties are altered -but differentially so- in MSNs from Q97 and from Q31 mice; excitatory and inhibitory synaptic currents exhibit higher amplitudes in Q31 but not Q97 MSNs, while excitatory synaptic currents occur at lower frequency in Q97 than in WT and Q31 MSNs; there is a reduced total dendritic length in Q31 -but not Q97- MSNs compared to WT, while spine density and number did not differ in MSNs in the three groups. The findings that Q31 MSNs differed from Q97 and WT neurons with regard to some physiological features and structurally suggest a novel role of the N17 domain in the function of WT Htt. The motor phenotype seen in Q97 mice was less robust than that reported in an earlier study (Gu et al., 2015), and the alterations to MSN physiological properties were largely consistent with changes reported previously in a number of other mouse models of HD. Together this study indicates that N17 plays a role in the modulation of the properties of MSNs in both mHtt and WT-Htt mice, but does not markedly exacerbate HD-like pathogenesis in the BACHD model.


Asunto(s)
Proteína Huntingtina/genética , Enfermedad de Huntington/genética , Potenciales de Acción , Animales , Cuerpo Estriado/patología , Cuerpo Estriado/fisiopatología , Dendritas/patología , Modelos Animales de Enfermedad , Potenciales Postsinápticos Excitadores , Femenino , Humanos , Proteína Huntingtina/química , Proteína Huntingtina/fisiología , Enfermedad de Huntington/patología , Enfermedad de Huntington/fisiopatología , Cojera Animal/genética , Cojera Animal/fisiopatología , Masculino , Ratones , Ratones Mutantes , Ratones Transgénicos , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/fisiología , Neuronas/patología , Neuronas/fisiología , Dominios Proteicos , Reflejo Anormal/genética , Reflejo Anormal/fisiología , Eliminación de Secuencia
9.
Elife ; 92020 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-32391794

RESUMEN

Vesicle fusion is mediated by assembly of SNARE proteins between opposing membranes. While previous work suggested an active role of SNARE transmembrane domains (TMDs) in promoting membrane merger (Dhara et al., 2016), the underlying mechanism remained elusive. Here, we show that naturally-occurring v-SNARE TMD variants differentially regulate fusion pore dynamics in mouse chromaffin cells, indicating TMD flexibility as a mechanistic determinant that facilitates transmitter release from differentially-sized vesicles. Membrane curvature-promoting phospholipids like lysophosphatidylcholine or oleic acid profoundly alter pore expansion and fully rescue the decelerated fusion kinetics of TMD-rigidifying VAMP2 mutants. Thus, v-SNARE TMDs and phospholipids cooperate in supporting membrane curvature at the fusion pore neck. Oppositely, slowing of pore kinetics by the SNARE-regulator complexin-2 withstands the curvature-driven speeding of fusion, indicating that pore evolution is tightly coupled to progressive SNARE complex formation. Collectively, TMD-mediated support of membrane curvature and SNARE force-generated membrane bending promote fusion pore formation and expansion.


Asunto(s)
Exocitosis , Fusión de Membrana , Complejos Multiproteicos/fisiología , Neurotransmisores/fisiología , Fosfolípidos/metabolismo , Proteínas SNARE/fisiología , Proteína 2 de Membrana Asociada a Vesículas/fisiología , Animales , Calcio/fisiología , Membrana Celular/metabolismo , Células Cultivadas , Células Cromafines , Cinética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Mutantes/fisiología , Unión Proteica , Dominios Proteicos , Vesículas Secretoras/fisiología
10.
Artículo en Inglés | MEDLINE | ID: mdl-32296391

RESUMEN

Although the rapid development of high-throughput sequencing has led to the identification of a large number of truncated or mutated steroid hormone receptor (SHR) variants, their clinical relevance remains to be defined. A platform for functional analysis of these SHR variants in cells would be instrumental for better assessing their impact on normal physiology and SHR-associated diseases. Here we have developed a new reporter system that allows rapid and accurate assessment of the transcriptional activity of SHR variants in cells. The reporter is a single construct containing a firefly luciferase reporter gene, whose expression is under the control of a promoter with multiple steroid hormone responsive elements, and a Renilla luciferase reporter gene, that is constitutively expressed under the control of an internal ribosome entry site (IRES) and is not regulated by steroid hormones. The corresponding SHR (wildtype or mutant/variant) is also expressed from the same construct. Using this improved reporter system, we revealed a large spectrum of transactivation activities within a set of previously identified mutations and variations of the androgen receptor (AR), the estrogen receptor α (ERα) and the glucocorticoid receptor (GR). This novel reporter system enables functional analysis of SHR mutants and variants in physiological and pathological settings, offering valuable preclinical, or diagnostic information for the understanding and treatment of associated diseases.


Asunto(s)
Bioensayo/métodos , Genes Reporteros , Vectores Genéticos/genética , Regiones Promotoras Genéticas/genética , Receptores Citoplasmáticos y Nucleares/fisiología , Activación Transcripcional/genética , Animales , Células COS , Células Cultivadas , Chlorocebus aethiops , Clonación Molecular/métodos , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/fisiología , Regulación de la Expresión Génica/efectos de los fármacos , Genes Reporteros/efectos de los fármacos , Células HEK293 , Células Hep G2 , Hormonas/farmacología , Humanos , Luciferasas de Luciérnaga/genética , Proteínas Mutantes/fisiología , Mutación , Regiones Promotoras Genéticas/efectos de los fármacos , Receptores Androgénicos/genética , Receptores Androgénicos/fisiología , Receptores Citoplasmáticos y Nucleares/genética , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/fisiología , Activación Transcripcional/efectos de los fármacos , Transfección/métodos
11.
Biomolecules ; 10(2)2020 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-32053881

RESUMEN

Pannexin 1 (Panx1) is a ubiquitously expressed hexameric integral membrane protein known to function as an adenosine triphosphate (ATP) release channel. Panx1 proteins exist in unglycosylated core form (Gly0). They undergo critical post-translational modifications forming the high mannose glycosylation state (Gly1) in the endoplasmic reticulum (ER) and the complex glycosylation state (Gly2) in the Golgi apparatus. The regulation of transition from the ER to the cell membrane is not fully understood. Using site-specific mutagenesis, dye uptake assays, and interaction testing, we identified two conserved aromatic residues, Trp123 and Tyr205, in the transmembrane domains 2 and 3 of the zebrafish panx1a protein. Results suggest that both residues primarily govern the assembly of panx1a subunits into channels, with mutant proteins failing to interact. The results provide insight into a mechanism enabling regulation of Panx1 oligomerization, glycosylation, and trafficking.


Asunto(s)
Aminoácidos Aromáticos/metabolismo , Conexinas/química , Conexinas/metabolismo , Proteínas de Pez Cebra/química , Proteínas de Pez Cebra/metabolismo , Pez Cebra/metabolismo , Aminoácidos Aromáticos/genética , Animales , Línea Celular Tumoral , Membrana Celular/metabolismo , Conexinas/genética , Retículo Endoplásmico/metabolismo , Glicosilación , Aparato de Golgi/metabolismo , Ratones , Mutagénesis Sitio-Dirigida , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Proteínas Mutantes/fisiología , Pliegue de Proteína , Multimerización de Proteína , Procesamiento Proteico-Postraduccional , Estabilidad Proteica , Transporte de Proteínas , Pez Cebra/genética , Proteínas de Pez Cebra/genética
12.
Biochim Biophys Acta Mol Cell Res ; 1867(1): 118573, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31678591

RESUMEN

Cytochrome c (Cyt c) released from mitochondria interacts with Apaf-1 to form the heptameric apoptosome, which initiates the caspase cascade to execute apoptosis. Although lysine residue at 72 (K72) of Cyt c plays an important role in the Cyt c-Apaf-1 interaction, the underlying mechanism of interaction between Cyt c and Apaf-1 is still not clearly defined. Here we identified multiple lysine residues including K72, which are also known to interact with ATP, to play a key role in Cyt c-Apaf-1 interaction. Mutation of these lysine residues abrogates the apoptosome formation causing inhibition of caspase activation. Using in-silico molecular docking, we have identified Cyt c-binding interface on Apaf-1. Although mutant Cyt c shows higher affinity for Apaf-1, the presence of Cyt c-WT restores the apoptosome activity. ATP addition modulates only mutant Cyt c binding to Apaf-1 but not WT Cyt c binding to Apaf-1. Using TCGA and cBioPortal, we identified multiple mutations in both Apaf-1 and Cyt c that are predicted to interfere with apoptosome assembly. We also demonstrate that transcript levels of various enzymes involved with dATP or ATP synthesis are increased in various cancers. Silencing of nucleotide metabolizing enzymes such as ribonucleotide reductase subunit M1 (RRM1) and ATP-producing glycolytic enzymes PKM2 attenuated ATP production and enhanced caspase activation. These findings suggest important role for lysine residues of Cyt c and nucleotides in the regulation of apoptosome-dependent apoptotic cell death as well as demonstrate how these mutations and nucleotides may have a pivotal role in human diseases such as cancer.


Asunto(s)
Apoptosomas/fisiología , Citocromos c/química , Simulación del Acoplamiento Molecular , Neoplasias/patología , Nucleótidos/química , Alanina/química , Alanina/genética , Sustitución de Aminoácidos , Apoptosomas/química , Factor Apoptótico 1 Activador de Proteasas/química , Factor Apoptótico 1 Activador de Proteasas/metabolismo , Estudios de Casos y Controles , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Células Cultivadas , Citocromos c/genética , Citocromos c/metabolismo , Femenino , Humanos , Lisina/química , Lisina/genética , Masculino , Modelos Moleculares , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Proteínas Mutantes/fisiología , Neoplasias/genética , Neoplasias/metabolismo , Nucleótidos/metabolismo , Células PC-3 , Unión Proteica/genética , Mapeo de Interacción de Proteínas , Multimerización de Proteína/genética , Transducción de Señal/genética
13.
J Transl Med ; 17(1): 351, 2019 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-31655630

RESUMEN

BACKGROUND: Biallelic PTPRQ pathogenic variants have been previously reported as causative for autosomal recessive non-syndromic hearing loss. In 2018 the first heterozygous PTPRQ variant has been implicated in the development of autosomal dominant non-syndromic hearing loss (ADNSHL) in a German family. The study presented the only, so far known, PTPRQ pathogenic variant (c.6881G>A) in ADNSHL. It is located in the last PTPRQ coding exon and introduces a premature stop codon (p.Trp2294*). METHODS: A five-generation Polish family with ADNSHL was recruited for the study (n = 14). Thorough audiological, neurotological and imaging studies were carried out to precisely define the phenotype. Genomic DNA was isolated from peripheral blood samples or buccal swabs of available family members. Clinical exome sequencing was conducted for the proband. Family segregation analysis of the identified variants was performed using Sanger sequencing. Single nucleotide polymorphism array on DNA samples from the Polish and the original German family was used for genome-wide linkage analysis. RESULTS: Combining clinical exome sequencing and family segregation analysis, we have identified the same (NM_001145026.2:c.6881G>A, NP_001138498.1:p.Trp2294*) PTPRQ alteration in the Polish ADNSHL family. Using genome-wide linkage analysis, we found that the studied family and the original German family derive from a common ancestor. Deep phenotyping of the affected individuals showed that in contrast to the recessive form, the PTPRQ-related ADNSHL is not associated with vestibular dysfunction. In both families ADNSHL was progressive, affected mainly high frequencies and had a variable age of onset. CONCLUSION: Our data provide the first confirmation of PTPRQ involvement in ADNSHL. The finding strongly reinforces the inclusion of PTPRQ to the small set of genes leading to both autosomal recessive and dominant hearing loss.


Asunto(s)
Pérdida Auditiva Sensorineural/genética , Proteínas Tirosina Fosfatasas Clase 3 Similares a Receptores/genética , Adolescente , Adulto , Edad de Inicio , Niño , Femenino , Genes Dominantes , Pérdida Auditiva Sensorineural/fisiopatología , Heterocigoto , Humanos , Masculino , Persona de Mediana Edad , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/fisiología , Mutación , Linaje , Terminación de la Cadena Péptídica Traduccional/genética , Fenotipo , Polonia , Polimorfismo de Nucleótido Simple , Proteínas Tirosina Fosfatasas Clase 3 Similares a Receptores/química , Proteínas Tirosina Fosfatasas Clase 3 Similares a Receptores/fisiología , Investigación Biomédica Traslacional , Adulto Joven
14.
J Biomol NMR ; 73(8-9): 493-507, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31407206

RESUMEN

Peptidyl Prolyl Isomerases (PPIases) accelerate cis-trans isomerization of prolyl peptide bonds. In rice, the PPIase LRT2 is essential for lateral root initiation. LRT2 displays in vitro isomerization of a highly conserved W-P peptide bond (104W-P105) in the natural substrate OsIAA11. OsIAA11 is a transcription repressor that, in response to the plant hormone auxin, is targeted to ubiquitin-mediated proteasomal degradation via specific recognition of the cis isomer of its 104W-P105 peptide bond. OsIAA11 controls transcription of specific genes, including its own, that are required for lateral root development. This auxin-responsive negative feedback circuit governs patterning and development of lateral roots along the primary root. The ability to tune LRT2 activity via mutagenesis is crucial for understanding and modeling the role of this bimodal switch in the auxin circuit and lateral root development. We present characterization of the thermal stability and isomerization rates of several LRT2 mutants acting on the OsIAA11 substrate. The thermally stable mutants display activities lower than that of wild-type (WT) LRT2. These include binding diminished but catalytically active P125K, binding incompetent W128A, and binding capable but catalytically incompetent H133Q mutations. Additionally, LRT2 homologs hCypA from human, TaCypA from Triticum aestivum (wheat) and PPIB from E. coli were shown to have 110, 50 and 60% of WT LRT2 activity on the OsIAA11 substrate. These studies identify several thermally stable LRT2 mutants with altered activities that will be useful for establishing relationships between cis-trans isomerization, auxin circuit dynamics, and lateral root development in rice.


Asunto(s)
Oryza/crecimiento & desarrollo , Proteínas de Plantas/química , Raíces de Plantas/crecimiento & desarrollo , Ácidos Indolacéticos , Isomerismo , Proteínas Mutantes/química , Proteínas Mutantes/fisiología , Resonancia Magnética Nuclear Biomolecular/métodos , Isomerasa de Peptidilprolil/química , Estabilidad Proteica
15.
J Cancer Res Clin Oncol ; 145(9): 2273-2283, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31428934

RESUMEN

OBJECTIVES: Recent research has classified lung adenocarcinoma patients with KRAS mutation into three subtypes by co-occurring genetic events in TP53 (KP subgroup), STK11/LKB1 (KL subgroup) and CDKN2A/B inactivation plus TTF-1 low expression (KC subgroup). The aim of this study was to identify valuable biomarkers by searching the candidate molecules that contribute to lung adenocarcinoma pathogenesis, especially KC subtype. MATERIALS AND METHODS: We analyzed the publicly available database and identified the candidate REG4 using the E-GEOD-31210 dataset, and then confirmed by TCGA dataset. In addition, an independent cohort of 55 clinical samples was analyzed by quantitative real-time PCR analysis. Functional studies and RNA sequencing were performed after silencing the REG4 expression. RESULTS: REG4, an important regulator of gastro-intestinal carcinogenesis, was highly expressed in KRAS mutant lung adenocarcinoma with low expression of TTF-1 (KC subtype). The results were validated both by gene expression analysis and immunohistochemistry study in an independent 55 clinical samples from Fudan University Shanghai Cancer Center. Further in vitro and in vivo functional assays revealed silencing REG4 expression significantly reduces cancer cell proliferation and tumorigenesis. Moreover, RNA sequencing and GSEA analysis displayed that REG4 knockdown might induce cell cycle arrest by regulating G2/M checkpoint and E2F targets. CONCLUSION: Our results indicate that REG4 plays an important role in KRAS-driven lung cancer pathogenesis and is a novel biomarker of lung adenocarcinoma subtype. Future studies are required to clarify the underlying mechanisms of REG4 in the division and proliferation of KC tumors and its potential therapeutic value.


Asunto(s)
Adenocarcinoma del Pulmón/diagnóstico , Biomarcadores de Tumor/genética , Transformación Celular Neoplásica/genética , Proteínas de Unión al ADN/genética , Neoplasias Pulmonares/diagnóstico , Proteínas Asociadas a Pancreatitis/genética , Proteínas Proto-Oncogénicas p21(ras)/fisiología , Factores de Transcripción/genética , Adenocarcinoma del Pulmón/genética , Adenocarcinoma del Pulmón/patología , Animales , Línea Celular Tumoral , Transformación Celular Neoplásica/patología , Estudios de Cohortes , Proteínas de Unión al ADN/metabolismo , Regulación hacia Abajo/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteínas Mutantes/genética , Proteínas Mutantes/fisiología , Proteínas Proto-Oncogénicas p21(ras)/genética , Factores de Transcripción/metabolismo
16.
Biochem Biophys Res Commun ; 515(2): 372-377, 2019 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-31155291

RESUMEN

Substitution of Ala for Thr residue in 155th position in γ-tropomyosin (Tpm3.12) is associated with muscle weakness. To understand the mechanisms of this defect, we studied the Ca2+-sensitivity of thin filaments in solution and multistep changes in mobility and spatial arrangement of actin, Tpm, and myosin heads during the ATPase cycle in reconstituted muscle fibres, using the polarized fluorescence microscopy. It was shown that the Ala155Thr (A155T) mutation increased the Ca2+-sensitivity of the thin filaments in solution. In the absence of the myosin heads in the muscle fibres, the mutation did not alter the ability of troponin to switch the thin filaments on and off at high and low Ca2+, respectively. However, upon the binding of myosin heads to the thin filaments at low Ca2+, the mutant Tpm was found to be markedly closer to the open position, than the wild-type Tpm. In the presence of the mutant Tpm, switching on of actin monomers and formation of the strong-binding state of the myosin heads were observed at low Ca2+, which indicated a higher myofilament Ca2+-sensitivity. The mutation decreased the amount of myosin heads bound strongly to actin at high Ca2+ and increased the number of these heads at relaxation. It is suggested that direct binding of myosin to Tpm may be one оf the reasons for muscle weakness associated with the A155T mutation. The use of reagents that decrease the Ca2+-sensitivity of the troponin complex may not be adequate to restore muscle function in patients with the A155T mutation.


Asunto(s)
Calcio/metabolismo , Debilidad Muscular/genética , Debilidad Muscular/fisiopatología , Tropomiosina/genética , Tropomiosina/fisiología , Actinas/metabolismo , Adenosina Trifosfatasas/metabolismo , Sustitución de Aminoácidos , Animales , Polarización de Fluorescencia , Humanos , Técnicas In Vitro , Masculino , Debilidad Muscular/etiología , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/fisiología , Mutación Missense , Miofibrillas/metabolismo , Subfragmentos de Miosina/metabolismo , Conejos , Tropomiosina/química , Troponina/metabolismo
17.
Hear Res ; 379: 79-88, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31103816

RESUMEN

Myosin VI is an actin-associated molecular motor vital for auditory and vestibular function. It is encoded by MYO6 located on chromosome 6q13 in human. Pathogenic variants in MYO6 have been associated with both dominant and recessive forms of hearing loss. However, the molecular mechanisms remain unclear. We established a humanized knock-in mouse model, Myo6-C442Y, to mimic the p.C442Y missense variant identified in human patients with autosomal dominant nonsyndromic hearing loss designated as DFNA22. We characterized hearing and inner ear morphologies of Myo6-C442Y and wild-type control mice. We found that both homozygous and heterozygous Myo6-C442Y mice exhibited hearing loss from three weeks after birth that rapidly progressed to profound deafness by six to nine weeks of age. The hearing loss corresponded to the degeneration of hair cells in the organ of Corti. We also observed disorganized stereocilia with irregular morphological features by immunohistochemistry and scanning electron microscopy. Additionally, hearing loss and inner-ear morphological anomalies were more pronounced and deteriorated more drastically in homozygous than in heterozygous Myo6-C442Y mice, indicating a semi-dominant inheritance pattern. Heterozygous Myo6-C442Y mice recapitulated the progressive postlingual sensorineural deafness in human, thus providing a useful model for elucidating the role myosin VI plays in the mammalian auditory system. Furthermore, the late-onset hearing loss of this mouse model may provide a therapeutic window for the emerging gene therapy, a promising strategy to treat certain forms of genetic deafness.


Asunto(s)
Pérdida Auditiva Sensorineural/genética , Proteínas Mutantes/genética , Cadenas Pesadas de Miosina/genética , Factores de Edad , Animales , Umbral Auditivo/fisiología , Modelos Animales de Enfermedad , Femenino , Técnicas de Sustitución del Gen , Genes Dominantes , Células Ciliadas Auditivas/patología , Pérdida Auditiva Sensorineural/patología , Pérdida Auditiva Sensorineural/fisiopatología , Heterocigoto , Homocigoto , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Transgénicos , Microscopía Electrónica de Rastreo , Proteínas Mutantes/fisiología , Mutación Missense , Cadenas Pesadas de Miosina/fisiología , Estereocilios/patología
18.
Cell Rep ; 27(1): 59-70.e4, 2019 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-30943415

RESUMEN

Neurodegenerative diseases commonly involve the disruption of circadian rhythms. Studies indicate that mutant Huntingtin (mHtt), the cause of Huntington's disease (HD), disrupts circadian rhythms often before motor symptoms are evident. Yet little is known about the molecular mechanisms by which mHtt impairs circadian rhythmicity and whether circadian clocks can modulate HD pathogenesis. To address this question, we used a Drosophila HD model. We found that both environmental and genetic perturbations of the circadian clock alter mHtt-mediated neurodegeneration. To identify potential genetic pathways that mediate these effects, we applied a behavioral platform to screen for clock-regulated HD suppressors, identifying a role for Heat Shock Protein 70/90 Organizing Protein (Hop). Hop knockdown paradoxically reduces mHtt aggregation and toxicity. These studies demonstrate a role for the circadian clock in a neurodegenerative disease model and reveal a clock-regulated molecular and cellular pathway that links clock function to neurodegenerative disease.


Asunto(s)
Relojes Circadianos/fisiología , Proteínas de Drosophila/metabolismo , Proteínas de Choque Térmico/metabolismo , Respuesta al Choque Térmico/fisiología , Proteína Huntingtina/metabolismo , Proteína Huntingtina/toxicidad , Agregación Patológica de Proteínas , Animales , Animales Modificados Genéticamente , Relojes Circadianos/genética , Modelos Animales de Enfermedad , Proteínas de Drosophila/genética , Drosophila melanogaster , Embrión no Mamífero , Femenino , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/fisiología , Proteína Huntingtina/genética , Enfermedad de Huntington/genética , Enfermedad de Huntington/metabolismo , Masculino , Proteínas Mutantes/fisiología , Agregación Patológica de Proteínas/genética , Agregación Patológica de Proteínas/metabolismo , Agregación Patológica de Proteínas/patología
19.
Hear Res ; 375: 14-24, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30831381

RESUMEN

Usher syndrome (USH) is the leading cause of inherited combined vision and hearing loss. However, mutations in most USH causative genes lead to other diseases, such as hearing loss only or vision loss only. The molecular mechanisms underlying the variable disease manifestations associated with USH gene mutations are unclear. This review focuses on an USH type 2 (USH2) gene encoding whirlin (WHRN; previously known as DFNB31), mutations in which have been found to cause either USH2 subtype USH2D or autosomal recessive non-syndromic deafness type 31 (DFNB31). This review summarizes the current knowledge about different whirlin isoforms encoded by WHRN orthologs in animal models, the interactions of different whirlin isoforms with their partners, and the function of whirlin isoforms in different cellular and subcellular locations. The recent findings regarding the function of whirlin isoforms suggest that disruption of different isoforms may be one of the mechanisms underlying the variable disease manifestations caused by USH gene mutations. This review also presents recent findings about the vestibular defects in Whrn mutant mouse models, which suggests that previous assumptions about the normal vestibular function of USH2 patients need to be re-evaluated. Finally, this review describes recent progress in developing therapeutics for diseases caused by WHRN mutations.


Asunto(s)
Pérdida Auditiva Sensorineural/genética , Pérdida Auditiva Sensorineural/fisiopatología , Proteínas de la Membrana/genética , Proteínas de la Membrana/fisiología , Síndromes de Usher/genética , Síndromes de Usher/fisiopatología , Animales , Cóclea/fisiopatología , Modelos Animales de Enfermedad , Terapia Genética , Células Ciliadas Auditivas/fisiología , Pérdida Auditiva Sensorineural/terapia , Humanos , Proteínas de la Membrana/química , Ratones , Proteínas Mutantes/genética , Proteínas Mutantes/fisiología , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiología , Retina/fisiopatología , Síndromes de Usher/terapia , Vestíbulo del Laberinto/fisiopatología
20.
J Bioinform Comput Biol ; 17(1): 1940003, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30866729

RESUMEN

A computation approach to identify the effect of missense mutations on the protein function is proposed. Using molecular dynamics simulation we have analyzed the gating kinetics of mutant NMDA synaptic receptors carrying mutations in their NR2 subunits. Analysis of channel geometry and Mg ion binding allowed to estimate the receptor conductivity. As a result, it was possible to identify the effect of these mutations on the generation of theta and gamma rhythms by the hippocampal neural network. Obtained results can be adapted for the analysis and evaluation of possible cognitive impairments caused by neurological diseases or consequences of radiation and other negative factors.


Asunto(s)
Hipocampo/fisiología , Modelos Neurológicos , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/fisiología , Sustitución de Aminoácidos , Animales , Región CA3 Hipocampal/citología , Región CA3 Hipocampal/fisiología , Biología Computacional , Simulación por Computador , Hipocampo/citología , Humanos , Potenciales de la Membrana , Simulación de Dinámica Molecular , Proteínas Mutantes/genética , Proteínas Mutantes/fisiología , Mutación Missense , Redes Neurales de la Computación , Subunidades de Proteína , Receptores de N-Metil-D-Aspartato/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...