Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Front Immunol ; 14: 1111172, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36926325

RESUMEN

Autoimmune bullous dermatoses (AIBD) are rare diseases that affect human skin and mucous membranes. Clinically, they are characterized by blister formation and/or erosions. Depending on the structures involved and the depth of blister formation, they are grouped into pemphigus diseases, pemphigoid diseases, and dermatitis herpetiformis. Classification of AIBD into their sub-entities is crucial to guide treatment decisions. One of the most sensitive screening methods for initial differentiation of AIBD is the indirect immunofluorescence (IIF) microscopy on tissue sections of monkey esophagus and primate salt-split skin, which are used to detect disease-specific autoantibodies. Interpretation of IIF patterns requires a detailed examination of the image by trained professionals automating this process is a challenging task with these highly complex tissue substrates, but offers the great advantage of an objective result. Here, we present computer-aided classification of esophagus and salt-split skin IIF images. We show how deep networks can be adapted to the specifics and challenges of IIF image analysis by incorporating segmentation of relevant regions into the prediction process, and demonstrate their high accuracy. Using this semi-automatic extension can reduce the workload of professionals when reading tissue sections in IIF testing. Furthermore, these results on highly complex tissue sections show that further integration of semi-automated workflows into the daily workflow of diagnostic laboratories is promising.


Asunto(s)
Enfermedades Autoinmunes , Penfigoide Ampolloso , Pénfigo , Enfermedades Cutáneas Vesiculoampollosas , Animales , Humanos , Técnica del Anticuerpo Fluorescente Indirecta/métodos , Vesícula , Enfermedades Autoinmunes/diagnóstico , Enfermedades Cutáneas Vesiculoampollosas/diagnóstico
2.
Aging Ment Health ; 25(3): 543-550, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-31941356

RESUMEN

OBJECTIVES: The current study examined how a technology system, "It's Never 2 Late" (iN2L), may help augment traditional rehabilitation strategies for older adults with dementia by improving engagement in therapy sessions and achieving better functional outcomes. METHOD: The study used a two group quasi-experimental design. Older adults with dementia (N = 96) were recruited from two rehabilitation departments housed within residential care communities. Participants received daily occupational and physical therapy sessions using treatment as usual (TAU) at one site (n = 49) or treatment with iN2L (n = 47) at the other site. A goal attainment approach was used to assess functional outcomes. It was hypothesized that patients whose therapists used iN2L in treatment will show greater attainment of therapy goals and greater engagement during OT and PT sessions than patients receiving TAU. It was also hypothesized that levels and improvement in engagement will mediate the association of treatment type (iN2L or TAU) with greater goal attainment. RESULTS: Participants in the iN2L treatment had significantly higher goal attainment than TAU, significantly higher levels of engagement at baseline, and significantly steeper increases in engagement over the course of therapy. The effects of treatment on goal attainment was significantly mediated by increases in engagement. CONCLUSION: Findings suggest that iN2L technology has the potential to increase treatment engagement and enhance rehabilitation outcomes among older adults with dementia.


Asunto(s)
Demencia , Calidad de Vida , Anciano , Humanos , Tecnología , Resultado del Tratamiento
3.
Annu Int Conf IEEE Eng Med Biol Soc ; 2019: 5886-5889, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31947189

RESUMEN

We propose a new area for wearable technology and interaction by acquiring gastrointestinal signals non-invasively from the abdomen. The mind-gut connection has flourished as a research area in the past two decades, elucidating the guts key role in stress, affect, and memory. Meanwhile, engineering advancements have shown potential in accuracy of non-invasive gastric recordings. Here, we investigate the design and specification of a wearable system for the recording of gut-brain activity non-invasively. We also present results from a preliminary pilot test of a wearable gut-brain computer interface (GBCI).


Asunto(s)
Interfaces Cerebro-Computador , Encéfalo/fisiología , Estómago/fisiología , Dispositivos Electrónicos Vestibles , Humanos
4.
Auto Immun Highlights ; 9(1): 8, 2018 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-30238164

RESUMEN

PURPOSE: Indirect immunofluorescence (IIF) on the human epithelial cell-line HEp-2 (or derivatives) serves as the gold standard in antinuclear antibody (ANA) screening. IIF, and its evaluation, is a labor-intensive method, making ANA testing a major challenge for present clinical laboratories. Nowadays, several automated ANA pattern recognition systems are on the market. In the current study, the EUROPattern Suite is evaluated for its use in daily practice in a routine setting. METHODS: A total of 1033 consecutive routine samples was used to screen for ANA. Results (positive/negative ANA screening, pattern identification and titer) were compared between software-generated results (EUROPattern) and visual interpretation (observer) of automatically acquired digital images. RESULTS: Considering the visual interpretation as reference, a relative sensitivity of 99.3% and a relative specificity of 88.9% were obtained for negative and positive discrimination by the software (EPa). A good agreement between visual and software-based interpretation was observed with respect to pattern recognition (mean kappa: for 7 patterns: 0.7). Interestingly, EPa software distinguished more patterns per positive sample than the observer (on average 1.5 and 1.2, respectively). Finally, a concordance of 99.3% was observed within the range of 1 titer step difference between EPa and observer. CONCLUSIONS: The ANA IIF results reported by the EPa software are in very good agreement with the results reported by the observer with respect to being negative/positive, pattern recognition and titer, making automated ANA IIF evaluation an objective and time-efficient tool for routine testing.

5.
Cytokine ; 83: 193-205, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27155818

RESUMEN

The promoter-intron-exon structure of genes evolve. While the structures of some IFN genes (e.g., piscine and amphibian Type I IFNs, most tetrapod IFN-λ genes) resemble those of other class II cytokines (e.g., interleukins-10, 19, 20, 22, 24, 26), the structures of other IFN genes differ significantly. Although all bony vertebrate IFN-γ genes lack the canonical third intron, and all amniote Type I IFN genes lack introns, only some IFN-λ genes lost their introns. Interestingly, these intronless IFN-λ genes are not preferentially related to one another nor are they clustered with canonical multi-intron IFN-λ genes. Hypothesizing that intronless IFN-λ genes repeatedly and independently evolved and transposed throughout the genome, we sought to understand the genetic processes involved in their intron loss and genomic migration. Utilizing the high conservation of the promoters, the UTRs and the ORFs of the IFN-λ genes, we collected data from two families of intronless IFN-λ genes, and developed a model supported by these data to explain how intronless IFN-λ genes evolved. (1) A cytoplasmic IFN-λ cDNA generated by reverse transcriptional activity enters the nucleus and attempts to recombine with its multi-exon progenitor. (2) Nuclear DNA synthesis at the 5' and 3' ends within recombination intermediates affixes the promoter onto the cDNA and preserves its 3' UTR. (3) Resolution of the recombination complex releases the promoter-associated cDNA. (4) The released intronless gene co-integrates with a highly duplicated sequence undergoing transposition. We propose that this process explains not only the evolution of the gene structure of IFN genes, but also the increased transposition of intronless genes in genomes, and may confer an evolutionary advantage.


Asunto(s)
Regiones no Traducidas 3' , ADN Complementario/genética , Evolución Molecular , Interferón gamma/genética , Intrones , Animales , Humanos
6.
J Immunol Res ; 2015: 742402, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26581239

RESUMEN

Systemic lupus erythematosus (SLE) is a severe rheumatic autoimmune disease with various clinical manifestations. Anti-dsDNA antibodies are an important immunological hallmark of SLE and their occurrence represents a major criterion for the diagnosis. Among the commonly applied test systems for determination of anti-dsDNA antibodies, the indirect immunofluorescence test (IIFT) using the flagellated kinetoplastida Crithidia luciliae is considered to be highly disease specific at moderate sensitivity. Since IIFT, however, is claimed to be affected by subjective interpretation and a lack of standardization, there has been an increasing demand for automated pattern interpretation of immunofluorescence reactions in recent years. Corresponding platforms are already available for evaluation of anti-nuclear antibody (ANA) IIFT on HEp-2 cells, the recommended "gold standard" for ANA screening in the diagnosis of various systemic rheumatic autoimmune diseases. For one of these systems, the "EUROPattern-Suite" computer-aided immunofluorescence microscopy (CAIFM), automated interpretation of microscopic fluorescence patterns was extended to the Crithidia luciliae based anti-dsDNA IIFT.


Asunto(s)
Anticuerpos Antinucleares/inmunología , Crithidia/inmunología , Técnica del Anticuerpo Fluorescente Indirecta , Automatización de Laboratorios , Humanos , Lupus Eritematoso Sistémico/diagnóstico , Lupus Eritematoso Sistémico/inmunología , Microscopía Fluorescente , Reproducibilidad de los Resultados , Enfermedades Reumáticas/diagnóstico , Enfermedades Reumáticas/inmunología , Sensibilidad y Especificidad , Flujo de Trabajo
7.
Cytokine ; 76(2): 480-495, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26228976

RESUMEN

Interferons (IFNs) are rapidly evolving cytokines released when viral infections are detected in cells. Previous research suggests that genes encoding IFNs and their receptors duplicated extensively throughout vertebrate evolution. We present molecular genetic evidence that supports the use of nonallelic homologous recombination (NAHR) to expand select IFN genes during amniote evolution. The duplication of long regions of genome (encompassing at least one functional IFN gene) followed by the insertion of this genome fragment near its parent's location, is commonly observed in many amniote genomes. Duplicates inserted away from duplication hotspots are not as frequently perturbed with new duplicates, and tend to survive long periods of evolution, sometimes becoming new IFN subtypes. Although most duplicates are inserted parallel to and near the original sequence, the insertion of the Kelch-like 9 gene within the Type I IFN locus of placental mammals promoted antiparallel insertion of gene duplicates between the Kelch-like 9 and IFN-ε loci. Genetic exchange between highly similar Type I gene duplicates as well as between Type III IFN gene duplicates homogenized their diversification. Oddly, Type III IFN genes migrated long distances throughout the genome more frequently than did Type I IFN genes. The inter-chromosomal movement of Type I IFN genes in amniotes correlated with complete intron loss in their gene structure, and repeatedly occurred with occasional Type III IFN genes.


Asunto(s)
Evolución Molecular , Interferones/genética , Animales , Femenino , Duplicación de Gen , Recombinación Homóloga , Humanos , Filogenia , Placenta/metabolismo , Embarazo , Primates/genética
8.
Cytokine ; 64(1): 286-97, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23830819

RESUMEN

Ectopic coexpression of the two chains of the Type I and Type III interferon (IFN) receptor complexes (IFN-αR1 and IFN-αR2c, or IFN-λR1 and IL-10R2) yielded sensitivity to IFN-alpha or IFN-lambda in only some cells. We found that IFN-αR1 and IFN-αR2c exhibit FRET only when expressed at equivalent and low levels. Expanded clonal cell lines expressing both IFN-αR1 and IFN-αR2c were sensitive to IFN-alpha only when IFN-αR1 and IFN-αR2c exhibited FRET in the absence of human IFN-alpha. Coexpression of RACK-1 or Jak1 enhanced the affinity of the interaction between IFN-αR1 and IFN-αR2c. Both IFN-αR1 and IFN-αR2c exhibited FRET with Jak1 and Tyk2. Together with data showing that disruption of the preassociation between the IFN-gamma receptor chains inhibited its biological activity, we propose that biologically active IFN receptors require ligand-independent juxtaposition of IFN receptor chains assisted by their associated cytosolic proteins.


Asunto(s)
Interferón-alfa/metabolismo , Interferón gamma/metabolismo , Receptor de Interferón alfa y beta/metabolismo , Receptores de Interferón/metabolismo , Línea Celular , Transferencia Resonante de Energía de Fluorescencia , Proteínas de Unión al GTP/metabolismo , Humanos , Janus Quinasa 1/metabolismo , Complejos Multiproteicos , Proteínas de Neoplasias/metabolismo , Unión Proteica , Receptores de Cinasa C Activada , Receptores de Superficie Celular/metabolismo , TYK2 Quinasa/metabolismo , Receptor de Interferón gamma
9.
Cytokine ; 64(1): 298-309, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23769803

RESUMEN

Experiments measuring Fluorescence Resonance Energy Transfer (FRET) between cytokine receptor chains and their associated proteins led to hypotheses describing their organization in intact cells. These interactions occur within a larger protein complex or within a given nano-environment. To illustrate this complexity empirically, we developed a protocol to analyze FRET among more than two fluorescent proteins (multi-FRET). In multi-FRET, we model FRET among more than two fluorophores as the sum of all possible pairwise interactions within the complex. We validated our assumption by demonstrating that FRET among pairs within a fluorescent triplet resembled FRET between each pair measured in the absence of the third fluorophore. FRET between two receptor chains increases with increasing FRET between the ligand-binding chain (e.g., IFN-γR1, IL-10R1 and IFN-λR1) and an acylated fluorescent protein that preferentially resides within subsections of the plasma membrane. The interaction of IL-10R2 with IFN-λR1 or IL-10R1 results in decreased FRET between IL-10R2 and the acylated fluorescent protein. Finally, we analyzed FRET among four fluorescent proteins to demonstrate that as FRET between IFN-γR1 and IFN-γR2 or between IFN-αR1 and IFN-αR2c increases, FRET among other pairs of proteins changes within each complex.


Asunto(s)
Transferencia Resonante de Energía de Fluorescencia/métodos , Receptor de Interferón alfa y beta/metabolismo , Receptores de Interferón/metabolismo , Receptores de Interleucina-10/metabolismo , Animales , Células COS , Línea Celular , Chlorocebus aethiops , Colorantes Fluorescentes , Células HEK293 , Humanos , Interferón gamma/metabolismo , Microdominios de Membrana/metabolismo , Microscopía Fluorescente , Complejos Multiproteicos/análisis , Unión Proteica , Receptor de Interferón gamma
10.
Cytokine ; 64(1): 272-85, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23796694

RESUMEN

The observed Fluorescence Resonance Energy Transfer (FRET) between fluorescently labeled proteins varies in cells. To understand how this variation affects our interpretation of how proteins interact in cells, we developed a protocol that mathematically separates donor-independent and donor-dependent excitations of acceptor, determines the electromagnetic interaction of donors and acceptors, and quantifies the efficiency of the interaction of donors and acceptors. By analyzing large populations of cells, we found that misbalanced or insufficient expression of acceptor or donor as well as their inefficient or reversible interaction influenced FRET efficiency in vivo. Use of red-shifted donors and acceptors gave spectra with less endogenous fluorescence but produced lower FRET efficiency, possibly caused by reduced quenching of red-shifted fluorophores in cells. Additionally, cryptic interactions between jellyfish FPs artefactually increased the apparent FRET efficiency. Our protocol can distinguish specific and nonspecific protein interactions even within highly constrained environments as plasma membranes. Overall, accurate FRET estimations in cells or within complex environments can be obtained by a combination of proper data analysis, study of sufficient numbers of cells, and use of properly empirically developed fluorescent proteins.


Asunto(s)
Transferencia Resonante de Energía de Fluorescencia/métodos , Quinasas Janus/análisis , Receptores de Interferón/análisis , Animales , Células COS , Línea Celular , Chlorocebus aethiops , Colorantes Fluorescentes/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Interferón gamma/metabolismo , Proteínas Luminiscentes/metabolismo , Microscopía Fluorescente/métodos , Complejos Multiproteicos , Receptores de Interferón/metabolismo , Coloración y Etiquetado , Receptor de Interferón gamma , Proteína Fluorescente Roja
11.
Clin Dev Immunol ; 2012: 651058, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23251220

RESUMEN

Indirect immunofluorescence (IIF) on human epithelial (HEp-2) cells is considered as the gold standard screening method for the detection of antinuclear autoantibodies (ANA). However, in terms of automation and standardization, it has not been able to keep pace with most other analytical techniques used in diagnostic laboratories. Although there are already some automation solutions for IIF incubation in the market, the automation of result evaluation is still in its infancy. Therefore, the EUROPattern Suite has been developed as a comprehensive automated processing and interpretation system for standardized and efficient ANA detection by HEp-2 cell-based IIF. In this study, the automated pattern recognition was compared to conventional visual interpretation in a total of 351 sera. In the discrimination of positive from negative samples, concordant results between visual and automated evaluation were obtained for 349 sera (99.4%, kappa = 0.984). The system missed out none of the 272 antibody-positive samples and identified 77 out of 79 visually negative samples (analytical sensitivity/specificity: 100%/97.5%). Moreover, 94.0% of all main antibody patterns were recognized correctly by the software. Owing to its performance characteristics, EUROPattern enables fast, objective, and economic IIF ANA analysis and has the potential to reduce intra- and interlaboratory variability.


Asunto(s)
Anticuerpos Antinucleares/química , Anticuerpos Antinucleares/inmunología , Células Epiteliales/química , Células Epiteliales/inmunología , Técnica del Anticuerpo Fluorescente Indirecta/métodos , Anticuerpos Antinucleares/sangre , Automatización de Laboratorios/métodos , Automatización de Laboratorios/normas , Línea Celular Tumoral , Técnica del Anticuerpo Fluorescente Indirecta/normas , Humanos , Estándares de Referencia , Sensibilidad y Especificidad
12.
Stem Cell Res Ther ; 2(2): 15, 2011 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-21401924

RESUMEN

INTRODUCTION: Local synthesis of interferon within B16 tumors mediates anti-tumor effects. Based on reports that stem cells are recruited to tumors, and because systemic administration of interferon causes dose-limiting undesirable side effects, we wanted to improve the anti-tumor effects of interferon while simultaneously minimizing its systemic side effects by employing mesenchymal stem cells (MSCs) as tumor-localized ectopic producers of interferon. Many vectors exist to fulfill this purpose, but their transfection efficiency and resulting expression levels vary considerably. METHODS: To follow both the recruitment to tumors and the synthesis of interferon by MSCs, we designed a bicistronic vector system that permits fluorescent visualization of vector-transfected and interferon-producing MSCs. We used Mu-IFNαA cDNA as the first cistron and the cherry fluorescent protein cDNA as the second cistron, whose translation requires the internal ribosome entry sequence (IRES) from the encephalomyocarditis virus 5' untranslated region. Observing inconsistent expression of these cistrons in various vectors and cell lines, especially compared with a control plasmid pmaxGFP, we optimized the expression of this bicistronic message by mutating pcDNA3 to facilitate exchange of the promoter and polyadenylation segments controlling both the gene of interest and the eukaryotic antibiotic resistance gene as well as the eukaryotic antibiotic resistance gene itself, and effectively compare the effects of these exchanges, creating plasmid pc3.5. RESULTS: Murine MSCs stably and ectopically expressing Mu-IFNαA inhibited the establishment of tumors in homogeneic C57/BL6 mice. Mu-IFNαA expressed from the bicistronic message is fully biologically active, but is expressed at only two-thirds of the level observed from a monocistronic message. Cap-dependent translation is threefold more efficient than IRES-driven translation in 293T, B16, and MSC cell lines. Both efficient expression and good transfection efficiency require strong expression of the gene of interest and a chimeric intron. High doses of Mu-IFNαA within tumors inhibited tumor establishment but may not inhibit tumor growth. CONCLUSIONS: Our modified vector and its derived plasmids will find use in stem cell therapeutics, gene expression, mRNA regulation, and transcription regulation. Local release of Mu-IFNαA within tumors may differently affect tumor establishment and tumor growth.


Asunto(s)
Células Madre Mesenquimatosas/metabolismo , Plásmidos/metabolismo , Regiones no Traducidas 5' , Animales , Línea Celular Tumoral , Virus de la Encefalomiocarditis/genética , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Células HEK293 , Humanos , Interferón-alfa/genética , Interferón-alfa/metabolismo , Células Madre Mesenquimatosas/citología , Ratones , Ratones Endogámicos C57BL , Neoplasias/prevención & control , Plásmidos/genética , Regiones Promotoras Genéticas , Transfección
13.
Mol Cell Biol ; 31(7): 1419-31, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21245386

RESUMEN

AUF1 is an AU-rich element (ARE)-binding protein that recruits translation initiation factors, molecular chaperones, and mRNA degradation enzymes to the ARE for mRNA destruction. We recently found chaperone Hsp27 to be an AUF1-associated ARE-binding protein required for tumor necrosis factor alpha (TNF-α) mRNA degradation in monocytes. Hsp27 is a multifunctional protein that participates in ubiquitination of proteins for their degradation by proteasomes. A variety of extracellular stimuli promote Hsp27 phosphorylation on three serine residues--Ser(15), Ser(78), and Ser(82)-by a number of kinases, including the mitogen-activated protein (MAP) pathway kinases p38 and MK2. Activating either kinase stabilizes ARE mRNAs. Likewise, ectopic expression of phosphomimetic mutant forms of Hsp27 stabilizes reporter ARE mRNAs. Here, we continued to examine the contributions of Hsp27 to mRNA degradation. As AUF1 is ubiquitinated and degraded by proteasomes, we addressed the hypothesis that Hsp27 phosphorylation controls AUF1 levels to modulate ARE mRNA degradation. Indeed, selected phosphomimetic mutants of Hsp27 promote proteolysis of AUF1 in a proteasome-dependent fashion and render ARE mRNAs more stable. Our results suggest that the p38 MAP kinase (MAPK)-MK2-Hsp27 signaling axis may target AUF1 destruction by proteasomes, thereby promoting ARE mRNA stabilization.


Asunto(s)
Proteínas de Choque Térmico HSP27/metabolismo , Ribonucleoproteína Heterogénea-Nuclear Grupo D/metabolismo , Procesamiento Proteico-Postraduccional , Estabilidad del ARN/genética , Secuencias Reguladoras de Ácido Ribonucleico/genética , Sustitución de Aminoácidos/genética , Línea Celular , Supervivencia Celular , Transferencia Resonante de Energía de Fluorescencia , Proteínas de Choque Térmico , Ribonucleoproteína Nuclear Heterogénea D0 , Humanos , Inmunoprecipitación , Interleucina-1beta/genética , Cinética , Chaperonas Moleculares , Proteínas Mutantes/metabolismo , Fosforilación , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica , Transducción de Señal , Factor de Necrosis Tumoral alfa/genética
14.
Mol Cell Biol ; 28(17): 5223-37, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18573886

RESUMEN

Controlled, transient cytokine production by monocytes depends heavily upon rapid mRNA degradation, conferred by 3' untranslated region-localized AU-rich elements (AREs) that associate with RNA-binding proteins. The ARE-binding protein AUF1 forms a complex with cap-dependent translation initiation factors and heat shock proteins to attract the mRNA degradation machinery. We refer to this protein assembly as the AUF1- and signal transduction-regulated complex, ASTRC. Rapid degradation of ARE-bearing mRNAs (ARE-mRNAs) requires ubiquitination of AUF1 and its destruction by proteasomes. Activation of monocytes by adhesion to capillary endothelium at sites of tissue damage and subsequent proinflammatory cytokine induction are prominent features of inflammation, and ARE-mRNA stabilization plays a critical role in the induction process. Here, we demonstrate activation-induced subunit rearrangements within ASTRC and identify chaperone Hsp27 as a novel subunit that is itself an ARE-binding protein essential for rapid ARE-mRNA degradation. As Hsp27 has well-characterized roles in protein ubiquitination as well as in adhesion-induced cytoskeletal remodeling and cell motility, its association with ASTRC may provide a sensing mechanism to couple proinflammatory cytokine induction with monocyte adhesion and motility.


Asunto(s)
Proteínas de Choque Térmico/metabolismo , Ribonucleoproteína Heterogénea-Nuclear Grupo D/metabolismo , Complejos Multiproteicos/metabolismo , Proteínas de Neoplasias/metabolismo , Subunidades de Proteína/metabolismo , Estabilidad del ARN , Secuencias Reguladoras de Ácido Ribonucleico/genética , Línea Celular Tumoral , Supervivencia Celular , Transferencia Resonante de Energía de Fluorescencia , Proteínas de Choque Térmico HSP27 , Ribonucleoproteína Nuclear Heterogénea D0 , Humanos , Chaperonas Moleculares , Unión Proteica , Factor de Necrosis Tumoral alfa/genética
15.
Cytokine Growth Factor Rev ; 18(5-6): 473-82, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17693124

RESUMEN

Interferons (IFNs) were discovered 50 years ago independently by Isaacs and Lindemann and by Nagata and Kojima. When it was later realized that IFNs are active at very low concentrations, research began to determine how their powerful effects were generated from such a small initial signal. It has since been established that interferons, as well as all other cytokines, employ cell surface receptors to translate their presence in the serum to a potent cellular response to a viral infection. These receptor complexes are composed of multiple distinct glycosylated transmembrane polypeptides, a number of protein tyrosine kinases, and interact transiently with a large variety of other proteins including transcription factors, phosphatases, signaling repressors, and adaptor proteins coupling the receptor to alternative signaling pathways. Three major receptor complexes exist that are exclusive to each of three major classes of interferon. Even though the effects of each major class of interferon vary physiologically, each receptor complex interacts with its ligand in similar ways and activates similar signaling cascades. In this mini-review, we take a historical perspective at the major events in the characterization of interferon receptors, discussing interesting results that still need to be explained.


Asunto(s)
Receptores de Interferón/historia , Animales , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Interferones/historia , Interferones/metabolismo , Receptores de Interferón/metabolismo , Investigación , Transducción de Señal
16.
Pharmacol Ther ; 113(1): 50-87, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17005254

RESUMEN

Protein arginine N-methylation is a post-translational modification whose influence on cell function is becoming widely appreciated. Protein arginine methyltransferases (PRMT) catalyze the methylation of terminal nitrogen atoms of guanidinium side chains within arginine residues of proteins. Recently, several new members of the PRMT family have been cloned and their catalytic function determined. In this report, we present a review and phylogenetic analysis of the PRMT found so far in genomes. PRMT are found in nearly all groups of eukaryotes. Many human PRMT originated early in eukaryote evolution. Homologs of PRMT1 and PRMT5 are found in nearly every eukaryote studied. The gene structure of PRMT vary: most introns appear to be inserted randomly into the open reading frame. The change in catalytic specificity of some PRMT occurred with changes in the arginine binding pocket within the active site. Because of the high degree of conservation of sequence among the family throughout evolution, creation of specific PRMT inhibitors in pathogenic organisms may be difficult, but could be very effective if developed. Furthermore, because of the intricate involvement of several PRMT in cellular physiology, their inhibition may be fraught with unwanted side effects. Nevertheless, development of pharmaceutical agents to control PRMT functions could lead to significant new targets.


Asunto(s)
Arginina/metabolismo , Evolución Molecular , Procesamiento Proteico-Postraduccional , Proteína-Arginina N-Metiltransferasas/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia Conservada , Inhibidores Enzimáticos/farmacología , Humanos , Isoenzimas , Metilación , Datos de Secuencia Molecular , Filogenia , Conformación Proteica , Proteína-Arginina N-Metiltransferasas/antagonistas & inhibidores , Proteína-Arginina N-Metiltransferasas/química , Proteína-Arginina N-Metiltransferasas/genética , Homología de Secuencia de Aminoácido , Especificidad por Sustrato
17.
Biochem Biophys Res Commun ; 342(2): 472-81, 2006 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-16487488

RESUMEN

We have identified a protein, FLJ12673 or FBXO11, that contains domains characteristically present in protein arginine methyltransferases (PRMTs). Immuno-purified protein expressed from one of the four splice variants in HeLa cells and in Escherichia coli exhibited methyltransferase activity. Monomethylarginine, symmetric, and asymmetric dimethylarginine (SDMA, ADMA) were formed on arginine residues. Accordingly, we have designated the protein PRMT9. PRMT9 is the third member of the PRMT family that forms SDMA modifications in proteins. Structurally, this protein is distinct from all other known PRMTs implying that convergent evolution allowed this protein to develop the ability to methylate arginine residues and evolved elements conserved in PRMTs to accomplish this.


Asunto(s)
Arginina/metabolismo , Proteínas F-Box/metabolismo , Proteína-Arginina N-Metiltransferasas/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Evolución Molecular , Proteínas F-Box/genética , Células HeLa , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , Metilación , Ratones , Datos de Secuencia Molecular , Familia de Multigenes , Proteína-Arginina N-Metiltransferasas/genética , Ratas , Alineación de Secuencia , Análisis de Secuencia de Proteína
18.
Cell Res ; 16(1): 55-69, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16467876

RESUMEN

We previously demonstrated using noninvasive technologies that the interferon-gamma (IFN-gamma) receptor complex is preassembled (1). In this report we determined how the receptor complex is preassembled and how the ligand-mediated conformational changes occur. The interaction of Stat1 with IFN-gammaR1 results in a conformational change localized to IFN-gammaR1. Jak1 but not Jak2 is required for the two chains of the IFN-gamma receptor complex (IFN-gammaR1 and IFN-gammaR2) to interact; however, the presence of both Jak1 and Jak2 is required to see any ligand-dependant conformational change. Two IFN-gammaR2 chains interact through species-specific determinants in their extracellular domains. Finally, these determinants also participate in the interaction of IFN-gammaR2 with IFN-gammaR1. These results agree with a detailed model of the IFN-gamma receptor that requires the receptor chains to be pre-associated constitutively for the receptor to be active.


Asunto(s)
Proteínas Tirosina Quinasas/fisiología , Proteínas Proto-Oncogénicas/fisiología , Receptores de Interferón/química , Receptores de Interferón/metabolismo , Factor de Transcripción STAT1/fisiología , Secuencia de Aminoácidos , Animales , Células COS , Chlorocebus aethiops , Humanos , Janus Quinasa 1 , Janus Quinasa 2 , Ligandos , Modelos Biológicos , Datos de Secuencia Molecular , Mutagénesis , Conformación Proteica , Receptores de Interferón/genética , Factor de Transcripción STAT1/metabolismo , Homología de Secuencia de Aminoácido , Transducción de Señal , Transfección , Receptor de Interferón gamma
19.
Cell Res ; 16(1): 113-23, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16467883

RESUMEN

The activation of Stat1 by the interferon-gamma (IFN-gamma) receptor complex is responsible for the transcription of a significant portion of IFN-gamma induced genes. Many of these genes are responsible for the induction of an apoptotic state in response to IFN-gamma. In the absence of Stat1 activation, IFN-gamma instead induces a proliferative response. Modifying Stat1 activation by IFN-gamma may have pharmacological benefits. We report that the rate of activation of Stat1 can be altered in HeLa cells by overexpressing either the IFN-gammaR1 chain or the IFN-gammaR2 chain. These alterations occur in hematopoietic cell lines: Raji cells and monocytic cell lines, which have average and above-average IFN-gammaR2 surface expression, activate Stat1 similarly to HeLa cells and HeLa cells overexpressing IFNgammaR2, respectively. The rapid Stat1 activation seen in HeLa cells can be inhibited by overexpressing a chimeric IFN-gammaR2 chain that does not bind Jak2 or (when high concentrations of IFN-gamma are used) by overexpressing IFN-gammaR1. These data are consistent with a model in which the recruitment of additional Jak2 activity to a signaling complex accelerates the rate of Stat1 activation. We conclude that the rate of activation of Stat1 in cells by IFN-gamma can be modified by regulating either receptor chain and speculate that pharmacological agents which modify receptor chain expression may alter IFN-gamma receptor signal transduction.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Receptores de Interferón/metabolismo , Receptores de Interferón/fisiología , Factor de Transcripción STAT1/metabolismo , Transcripción Genética , Línea Celular , Células HeLa , Humanos , Interferón gamma/farmacología , Factor de Transcripción STAT1/fisiología , Transducción de Señal , Transactivadores , Transfección , Receptor de Interferón gamma
20.
Biochem Biophys Res Commun ; 340(2): 377-85, 2006 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-16364239

RESUMEN

We used fluorescence resonance energy transfer previously to show that the interferon-gamma (IFN-gamma) receptor complex is a preformed entity mediated by constitutive interactions between the IFN-gammaR2 and IFN-gammaR1 chains, and that this preassembled entity changes its structure after the treatment of cells with IFN-gamma. We applied this technique to determine the structure of the interleukin-10 (IL-10) receptor complex and whether it undergoes a similar conformational change after treatment of cells with IL-10. We report that, like the IFN-gamma receptor complex, the IL-10 receptor complex is preassembled: constitutive but weaker interactions occur between the IL-10R1 and IL-10R2 chains, and between two IL-10R2 chains. The IL-10 receptor complex undergoes no major conformational changes when cells are treated with cellular or Epstein-Barr viral IL-10. Receptor complex preassembly may be an inherent feature of Class 2 cytokine receptor complexes.


Asunto(s)
Interleucina-10/metabolismo , Subunidades de Proteína/metabolismo , Receptores de Interleucina/metabolismo , Animales , Células COS , Membrana Celular/metabolismo , Chlorocebus aethiops , Transferencia Resonante de Energía de Fluorescencia , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Receptores de Interferón/química , Receptores de Interferón/metabolismo , Receptores de Interleucina/química , Receptores de Interleucina-10 , Receptor de Interferón gamma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...