Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Vaccine ; 38(48): 7569-7577, 2020 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-33071001

RESUMEN

Recent malaria vaccine trials in endemic areas have yielded disparate results compared to studies conducted in non-endemic areas. A workshop was organized to discuss the differential pre-erythrocytic stage malaria vaccine (Pre-E-Vac) efficacies and underlying protective immunity under various conditions. It was concluded that many factors, including vaccine technology platforms, host genetics or physiologic conditions, and parasite and mosquito vector variations, may all contribute to Pre-E-Vac efficacy. Cross-disciplinary approaches are needed to decipher the multi-dimensional variables that contribute to the observed vaccine hypo-responsiveness. The malaria vaccine community has an opportunity to leverage recent advances in immunology, systems vaccinology, and high dimensionality data science methodologies to generate new clinical datasets with unprecedented levels of functional resolution as well as capitalize on existing datasets for comprehensive and aggregate analyses. These approaches would help to unlock our understanding of Pre-E-Vac immunology and to translate new candidates from the laboratory to the field more predictably.


Asunto(s)
Eritrocitos , Vacunas contra la Malaria , Malaria , Animales , Culicidae , Vectores de Enfermedades , Eritrocitos/inmunología , Malaria/epidemiología , Malaria/prevención & control
2.
Appl Environ Microbiol ; 75(7): 2027-36, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19201967

RESUMEN

Varicella-zoster virus (VZV; human herpesvirus 3) is the etiological cause of chickenpox and, upon reactivation from latency, zoster. Currently, vaccines are available to prevent both diseases effectively. A critical requirement for the manufacturing of safe and potent vaccines is the measurement of the biological activity to ensure proper dosing and efficacy, while minimizing potentially harmful secondary effects induced by immunization. In the case of live virus-containing vaccines, such as VZV-containing vaccines, biological activity is determined using an infectivity assay in a susceptible cellular host in vitro. Infectivity measurements generally rely on the enumeration of plaques by visual inspection of an infected cell monolayer. These plaque assays are generally very tedious and labor intensive and have modest throughput and high associated variability. In this study, we have developed a flow cytometry assay to measure the infectivity of the attenuated vaccine strain (vOka/Merck) of VZV in MRC-5 cells with improved throughput. The assay is performed in 96-well tissue culture microtiter plates and is based on the detection and quantification of infected cells expressing VZV glycoproteins on their surfaces. Multiple assay parameters have been investigated, including specificity, limit of detection, limit of quantification, range of linear response, signal-to-noise ratio, and precision. This novel assay appears to be in good concordance with the classical plaque assay results and therefore provides a viable, higher-throughput alternative to the plaque assay.


Asunto(s)
Automatización/métodos , Vacuna contra la Varicela , Citometría de Flujo/métodos , Herpesvirus Humano 3/aislamiento & purificación , Línea Celular , Humanos , Sensibilidad y Especificidad , Ensayo de Placa Viral
3.
Trends Biochem Sci ; 27(1): 19-26, 2002 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11796220

RESUMEN

Ligands of the TNF (tumour necrosis factor) superfamily have pivotal roles in the organization and function of the immune system, and are implicated in the aetiology of several acquired and genetic diseases. TNF ligands share a common structural motif, the TNF homology domain (THD), which binds to cysteine-rich domains (CRDs) of TNF receptors. CRDs are composed of structural modules, whose variation in number and type confers heterogeneity upon the family. Protein folds reminiscent of the THD and CRD are also found in other protein families, raising the possibility that the mode of interaction between TNF and TNF receptors might be conserved in other contexts.


Asunto(s)
Factor de Necrosis Tumoral alfa/química , Secuencia de Aminoácidos , Ligandos , Sustancias Macromoleculares , Datos de Secuencia Molecular , Estructura Molecular , Conformación Proteica , Pliegue de Proteína , Receptores del Factor de Necrosis Tumoral/química , Homología de Secuencia de Aminoácido
4.
Curr Biol ; 12(10): 838-43, 2002 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-12015121

RESUMEN

Apoptotic cell death is characterized by several morphological nuclear changes, such as chromatin condensation and extensive fragmentation of chromosomal DNA. These alterations are primarily triggered through the activation of caspases, which subsequently cleave nuclear substrates. Caspase-3 induces processing of Acinus, which leads to chromatin condensation. DNA fragmentation is dependent on the DNase CAD, which is released from its inhibitor, ICAD, upon cleavage by caspase-3. DNA degradation is also induced by AIF and endonuclease G, which are both released from mitochondria upon death stimuli but do not require prior processing by caspases for their DNase activity. Here we report the identification of a widely expressed helicase designated Helicard, which contains two N-terminal CARD domains and a C-terminal helicase domain. Upon apoptotic stimuli, Helicard is cleaved by caspases, thereby separating the CARD domains from the helicase domain. While Helicard localizes in the cytoplasm, the helicase-containing fragment is found in the nucleus. Helicard accelerates Fas ligand-mediated DNA degradation, whereas a noncleavable or a helicase-dead Helicard mutant does not, implicating Helicard in the nuclear remodeling occurring during apoptosis.


Asunto(s)
Apoptosis , Fragmentación del ADN , ADN Helicasas/química , ADN Helicasas/metabolismo , Secuencia de Aminoácidos , Animales , Caspasa 3 , Caspasas/metabolismo , ADN Helicasas/genética , Perfilación de la Expresión Génica , Células HeLa , Humanos , Ratones , Datos de Secuencia Molecular , Especificidad de Órganos , Reacción en Cadena de la Polimerasa , Estructura Terciaria de Proteína , Ratas , Homología de Secuencia de Aminoácido , Transfección
5.
PLoS One ; 12(1): e0170640, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28125650

RESUMEN

Clostridium difficile infections (CDI) are a leading cause of nosocomial diarrhea in the developed world. The main virulence factors of the bacterium are the large clostridial toxins (LCTs), TcdA and TcdB, which are largely responsible for the symptoms of the disease. Recent outbreaks of CDI have been associated with the emergence of hypervirulent strains, such as NAP1/BI/027, many strains of which also produce a third toxin, binary toxin (CDTa and CDTb). These hypervirulent strains have been associated with increased morbidity and higher mortality. Here we present pre-clinical data describing a novel tetravalent vaccine composed of attenuated forms of TcdA, TcdB and binary toxin components CDTa and CDTb. We demonstrate, using the Syrian golden hamster model of CDI, that the inclusion of binary toxin components CDTa and CDTb significantly improves the efficacy of the vaccine against challenge with NAP1 strains in comparison to vaccines containing only TcdA and TcdB antigens, while providing comparable efficacy against challenge with the prototypic, non-epidemic strain VPI10463. This combination vaccine elicits high neutralizing antibody titers against TcdA, TcdB and binary toxin in both hamsters and rhesus macaques. Finally we present data that binary toxin alone can act as a virulence factor in animal models. Taken together, these data strongly support the inclusion of binary toxin in a vaccine against CDI to provide enhanced protection from epidemic strains of C. difficile.


Asunto(s)
Toxinas Bacterianas/genética , Vacunas Bacterianas/administración & dosificación , Infecciones por Clostridium/prevención & control , Enterotoxinas/genética , Animales , Toxinas Bacterianas/toxicidad , Vacunas Bacterianas/genética , Clostridioides difficile/efectos de los fármacos , Clostridioides difficile/patogenicidad , Infecciones por Clostridium/genética , Infecciones por Clostridium/microbiología , Cricetinae , Modelos Animales de Enfermedad , Enterotoxinas/toxicidad , Humanos , Macaca mulatta/microbiología , Mesocricetus/microbiología
6.
Methods Mol Biol ; 1403: 385-96, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27076142

RESUMEN

Clostridium difficile is a gram-positive bacterium responsible for a large proportion of nosocomial infections in the developed world. C. difficile secretes toxins A and B (TcdA and TcdB) and both toxins act synergistically to induce a spectrum of pathological responses in infected individuals ranging from pseudomembranous colitis to C. difficile-associated diarrhea. Toxins A and B have been actively investigated as components of prophylactic vaccine as well as targets for therapeutic intervention with antibodies. Expression of such toxins by recombinant technology is often difficult and may require special handling and adherence to strict safety regulations during the manufacturing process due to the inherent toxicity of the proteins. Both toxins are large proteins (308 kDa and 270 kDa, respectively) and contain distinct domains mediating cell attachment, cellular translocation, and enzymatic (glucosidase) activity. Here we describe methods to produce fragments of Toxin B for their subsequent evaluation as components of experimental C. difficile vaccines. Methods presented include selection of fragments encompassing distinct functional regions of Toxin B, purification methods to yield high quality proteins, and analytical evaluation techniques. The approach presented focuses on Toxin B but could be applied to the other component, Toxin A, and/or to any difficult to express or toxic protein.


Asunto(s)
Vacunas Bacterianas/inmunología , Infecciones por Clostridium/prevención & control , Animales , Antígenos Bacterianos/inmunología , Clostridioides difficile/inmunología , Infecciones por Clostridium/inmunología , Diseño de Fármacos , Humanos , Vacunas de Subunidad/inmunología
7.
Oncogene ; 21(39): 6132-7, 2002 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-12203125

RESUMEN

Death induced by doxorubicin (dox) in neuroblastoma (NB) cells was originally thought to occur via the Fas pathway, however since studies suggest that caspase-8 expression is silenced in most high stage NB tumors, it is more probable that dox-induced death occurs via a different mechanism. Caspase-8 silenced N-type invasive NB cell lines LAN-1 and IMR-32 were investigated for their sensitivity to dox, and compared to S-type noninvasive SH-EP NB cells expressing caspase-8. All cell lines had similar sensitivities to dox, independently of caspase-8 expression. Dox induced caspase-3, -7, -8 and -9 and Bid cleavage in S-type cells and death was blocked by caspase inhibitors but not by oxygen radical scavenger BHA. In contrast, dox-induced death in N-type cells was caspase-independent and was inhibited by BHA. Dox induced a drop in mitochondrial membrane permeability in all cell lines. Dox-induced death in S-type cells gave rise to apoptotic nuclei, whereas in N-type cells nuclei were non-apoptotic in morphology. Transfection of SH-EP cells with a dominant negative FADD mutant inhibited TRAIL-induced death, but had no effect on dox-induced apoptosis. These results suggest that S-type cells undergo apoptosis after dox treatment independently of death receptors, whereas N-type cells are killed by a caspase-independent mechanism.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Proteínas Portadoras/metabolismo , Caspasas/metabolismo , Doxorrubicina/farmacología , Neuroblastoma/tratamiento farmacológico , Neuroblastoma/metabolismo , Antioxidantes/farmacología , Apoptosis/fisiología , Proteínas Reguladoras de la Apoptosis , Hidroxianisol Butilado/farmacología , Proteínas Portadoras/genética , Proteínas Portadoras/inmunología , Inhibidores de Caspasas , Núcleo Celular/metabolismo , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , Resistencia a Antineoplásicos/fisiología , Inhibidores Enzimáticos/farmacología , Proteína de Dominio de Muerte Asociada a Fas , Genes Dominantes , Humanos , Glicoproteínas de Membrana/farmacología , Invasividad Neoplásica/fisiopatología , Neuroblastoma/patología , Especies Reactivas de Oxígeno , Ligando Inductor de Apoptosis Relacionado con TNF , Transfección , Factor de Necrosis Tumoral alfa/farmacología , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
8.
Vaccine ; 33(52): 7496-505, 2015 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-26428458

RESUMEN

Until recently, malaria vaccine development efforts have focused almost exclusively on a handful of well characterized Plasmodium falciparum antigens. Despite dedicated work by many researchers on different continents spanning more than half a century, a successful malaria vaccine remains elusive. Sequencing of the P. falciparum genome has revealed more than five thousand genes, providing the foundation for systematic approaches to discover candidate vaccine antigens. We are taking advantage of this wealth of information to discover new antigens that may be more effective vaccine targets. Herein, we describe different approaches to large-scale screening of the P. falciparum genome to identify targets of either antibody responses or T cell responses using human specimens collected in Controlled Human Malaria Infections (CHMI) or under conditions of natural exposure in the field. These genome, proteome and transcriptome based approaches offer enormous potential for the development of an efficacious malaria vaccine.


Asunto(s)
Antígenos de Protozoos/inmunología , Genoma de Protozoos , Vacunas contra la Malaria , Malaria Falciparum/inmunología , Malaria Falciparum/prevención & control , Plasmodium falciparum/genética , Plasmodium falciparum/inmunología , Anticuerpos Antiprotozoarios/biosíntesis , Anticuerpos Antiprotozoarios/inmunología , Antígenos de Protozoos/química , Genómica , Ensayos Analíticos de Alto Rendimiento , Humanos , Inmunidad Innata , Vacunas contra la Malaria/inmunología , Proteoma/inmunología , Transcriptoma
9.
Clin Vaccine Immunol ; 22(11): 1206-18, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26446421

RESUMEN

Chlamydia trachomatis is the causative agent of the most frequently reported bacterial sexually transmitted infection, the total burden of which is underestimated due to the asymptomatic nature of the infection. Untreated C. trachomatis infections can cause significant morbidities, including pelvic inflammatory disease and tubal factor infertility (TFI). The human immune response against C. trachomatis, an obligate intracellular bacterium, is poorly characterized but is thought to rely on cell-mediated immunity, with CD4(+) and CD8(+) T cells implicated in protection. In this report, we present immune profiling data of subjects enrolled in a multicenter study of C. trachomatis genital infection. CD4(+) and CD8(+) T cells from subjects grouped into disease-specific cohorts were screened using a C. trachomatis proteomic library to identify the antigen specificities of recall T cell responses after natural exposure by measuring interferon gamma (IFN-γ) levels. We identified specific T cell responses associated with the resolution of infection, including unique antigens identified in subjects who spontaneously cleared infection and different antigens associated with C. trachomatis-related sequelae, such as TFI. These data suggest that novel and unique C. trachomatis T cell antigens identified in individuals with effective immune responses can be considered as targets for vaccine development, and by excluding antigens associated with deleterious sequelae, immune-mediated pathologies may be circumvented.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Infecciones por Chlamydia/inmunología , Chlamydia trachomatis/inmunología , Infecciones del Sistema Genital/inmunología , Adolescente , Adulto , Anticuerpos Antibacterianos/inmunología , Antígenos Bacterianos/inmunología , Infecciones por Chlamydia/microbiología , Chlamydia trachomatis/genética , Epítopos de Linfocito T/inmunología , Femenino , Humanos , Inmunidad Celular , Interferón gamma/inmunología , Masculino , Persona de Mediana Edad , Proteómica , Infecciones del Sistema Genital/microbiología , Adulto Joven
10.
ISME J ; 9(2): 321-32, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25036923

RESUMEN

Clostridium difficile infections (CDI) are caused by colonization and growth of toxigenic strains of C. difficile in individuals whose intestinal microbiota has been perturbed, in most cases following antimicrobial therapy. Determination of the protective commensal gut community members could inform the development of treatments for CDI. Here, we utilized the lethal enterocolitis model in Syrian golden hamsters to analyze the microbiota disruption and recovery along a 20-day period following a single dose of clindamycin on day 0, inducing in vivo susceptibility to C. difficile infection. To determine susceptibility in vitro, spores of strain VPI 10463 were cultured with and without soluble hamster fecal filtrates and growth was quantified by quantitative PCR and toxin immunoassay. Fecal microbial population changes over time were tracked by 16S ribosomal RNA gene analysis via V4 sequencing and the PhyloChip assay. C. difficile culture growth and toxin production were inhibited by the presence of fecal extracts from untreated hamsters but not extracts collected 5 days post-administration of clindamycin. In vitro inhibition was re-established by day 15, which correlated with resistance of animals to lethal challenge. A substantial fecal microbiota shift in hamsters treated with antibiotics was observed, marked by significant changes across multiple phyla including Bacteroidetes and Proteobacteria. An incomplete return towards the baseline microbiome occurred by day 15 correlating with the inhibition of C. difficile growth in vitro and in vivo. These data suggest that soluble factors produced by the gut microbiota may be responsible for the suppression of C. difficile growth and toxin production.


Asunto(s)
Clostridioides difficile , Infecciones por Clostridium/microbiología , Colon/microbiología , Microbiota , Animales , Antibacterianos/farmacología , Clindamicina/farmacología , Clostridioides difficile/clasificación , Clostridioides difficile/efectos de los fármacos , Clostridioides difficile/crecimiento & desarrollo , Cricetinae , Enterocolitis/microbiología , Heces/microbiología , Masculino , Mesocricetus , Modelos Biológicos
11.
FEBS Lett ; 527(1-3): 250-4, 2002 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-12220669

RESUMEN

Death effector domains (DEDs) are protein-protein interaction domains found in the death inducing signaling complex (DISC). Performing a structure-based alignment of all DED sequences we identified a region of high diversity in alpha-helix 3 and propose a classification of DEDs into class I DEDs typically containing a stretch of basic residues in the alpha-helix 3 region whereas DEDs of class II do not. Functional assays using mutants of Fas-associated death domain revealed that this basic region influences binding and recruitment of caspase-8 and cellular FLICE inhibitor protein to the DISC.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Apoptosis/fisiología , Proteínas Portadoras/química , Proteínas Portadoras/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Secuencia de Aminoácidos , Sitios de Unión , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD , Proteínas Portadoras/genética , Caspasa 3 , Caspasas/metabolismo , Células Cultivadas , Proteína de Dominio de Muerte Asociada a Fas , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Mutación , Conformación Proteica , Estructura Terciaria de Proteína , Transducción de Señal
12.
Vaccine ; 32(24): 2812-8, 2014 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-24662701

RESUMEN

Clostridium difficile infection (CDI) is the major cause of antibiotic-associated diarrhea and pseudomembranous colitis, a disease associated with significant morbidity and mortality. The disease is mostly of nosocomial origin, with elderly patients undergoing anti-microbial therapy being particularly at risk. C. difficile produces two large toxins: Toxin A (TcdA) and Toxin B (TcdB). The two toxins act synergistically to damage and impair the colonic epithelium, and are primarily responsible for the pathogenesis associated with CDI. The feasibility of toxin-based vaccination against C. difficile is being vigorously investigated. A vaccine based on formaldehyde-inactivated Toxin A and Toxin B (toxoids) was reported to be safe and immunogenic in healthy volunteers and is now undergoing evaluation in clinical efficacy trials. In order to eliminate cytotoxic effects, a chemical inactivation step must be included in the manufacturing process of this toxin-based vaccine. In addition, the large-scale production of highly toxic antigens could be a challenging and costly process. Vaccines based on non-toxic fragments of genetically engineered versions of the toxins alleviate most of these limitations. We have evaluated a vaccine assembled from two recombinant fragments of TcdB and explored their potential as components of a novel experimental vaccine against CDI. Golden Syrian hamsters vaccinated with recombinant fragments of TcdB combined with full length TcdA (Toxoid A) developed high titer IgG responses and potent neutralizing antibody titers. We also show here that the recombinant vaccine protected animals against lethal challenge with C. difficile spores, with efficacy equivalent to the toxoid vaccine. The development of a two-segment recombinant vaccine could provide several advantages over toxoid TcdA/TcdB such as improvements in manufacturability.


Asunto(s)
Proteínas Bacterianas/inmunología , Toxinas Bacterianas/inmunología , Vacunas Bacterianas/inmunología , Infecciones por Clostridium/prevención & control , Enterocolitis Seudomembranosa/prevención & control , Enterotoxinas/inmunología , Animales , Anticuerpos Antibacterianos/sangre , Anticuerpos Neutralizantes/sangre , Clostridioides difficile , Inmunoglobulina G/sangre , Masculino , Mesocricetus , Pruebas de Neutralización , Proteínas Recombinantes/inmunología , Vacunas Sintéticas/inmunología
13.
Clin Vaccine Immunol ; 20(4): 517-25, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23389929

RESUMEN

Clostridium difficile produces two major virulence toxins, toxin A (TcdA) and toxin B (TcdB). Antitoxin antibodies, especially neutralizing antibodies, have been shown to be associated with a lower incidence of C. difficile infection (CDI) recurrence, and antibody levels are predictive of asymptomatic colonization. The development of an assay to detect the presence of neutralizing antibodies in animal and human sera for the evaluation of vaccine efficacy is highly desired. We have developed such an assay, which allows for the quantification of the effect of toxins on eukaryotic cells in an automated manner. We describe here the optimization of this assay to measure toxin potency as well as neutralizing antibody (NAb) activity against C. difficile toxins using a design-of-experiment (DOE) methodology. Toxin concentration and source, cell seeding density, and serum-toxin preincubation time were optimized in the assay using Vero cells. The assay was shown to be robust and to produce linear results across a range of antibody concentrations. It can be used to quantify neutralizing antibodies in sera of monkeys and hamsters immunized with C. difficile toxoid vaccines. This assay was shown to correlate strongly with traditional assays which rely on labor-intensive methods of determining neutralizing antibody titers by visual microscopic inspection of intoxicated-cell monolayers. This assay has utility for the selection and optimization of C. difficile vaccine candidates.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Proteínas Bacterianas/inmunología , Toxinas Bacterianas/inmunología , Clostridioides difficile/inmunología , Técnicas Citológicas/métodos , Enterotoxinas/inmunología , Pruebas de Neutralización/métodos , Proteínas Represoras/inmunología , Animales , Automatización de Laboratorios/métodos , Chlorocebus aethiops , Cricetinae , Masculino , Mesocricetus , Células Vero
14.
J Virol Methods ; 166(1-2): 1-11, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20117140

RESUMEN

Vaccine manufacturing requires constant analytical monitoring to ensure reliable quality and a consistent safety profile of the final product. Concentration and bioactivity of active components of the vaccine are key attributes routinely evaluated throughout the manufacturing cycle and for product release and dosage. In the case of live attenuated virus vaccines, bioactivity is traditionally measured in vitro by infection of susceptible cells with the vaccine followed by quantification of virus replication, cytopathology or expression of viral markers. These assays are typically multi-day procedures that require trained technicians and constant attention. Considering the need for high volumes of testing, automation and streamlining of these assays is highly desirable. In this study, the automation and streamlining of a complex infectivity assay for Varicella Zoster Virus (VZV) containing test articles is presented. The automation procedure was completed using existing liquid handling infrastructure in a modular fashion, limiting custom-designed elements to a minimum to facilitate transposition. In addition, cellular senescence data provided an optimal population doubling range for long term, reliable assay operation at high throughput. The results presented in this study demonstrate a successful automation paradigm resulting in an eightfold increase in throughput while maintaining assay performance characteristics comparable to the original assay.


Asunto(s)
Automatización , Vacuna contra la Varicela/efectos adversos , Herpesvirus Humano 3/patogenicidad , Virología/métodos , Línea Celular , Humanos , Control de Calidad , Vacunas Atenuadas/efectos adversos , Cultivo de Virus/métodos
15.
J Biol Chem ; 281(20): 13964-71, 2006 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-16547002

RESUMEN

Ligands of the tumor necrosis factor superfamily (TNFSF) (4-1BBL, APRIL, BAFF, CD27L, CD30L, CD40L, EDA1, EDA2, FasL, GITRL, LIGHT, lymphotoxin alpha, lymphotoxin alphabeta, OX40L, RANKL, TL1A, TNF, TWEAK, and TRAIL) bind members of the TNF receptor superfamily (TNFRSF). A comprehensive survey of ligand-receptor interactions was performed using a flow cytometry-based assay. All ligands engaged between one and five receptors, whereas most receptors only bound one to three ligands. The receptors DR6, RELT, TROY, NGFR, and mouse TNFRH3 did not interact with any of the known TNFSF ligands, suggesting that they either bind other types of ligands, function in a ligand-independent manner, or bind ligands that remain to be identified. The study revealed that ligand-receptor pairs are either cross-reactive between human and mouse (e.g. Tweak/Fn14, RANK/RANKL), strictly species-specific (GITR/GITRL), or partially species-specific (e.g. OX40/OX40L, CD40/CD40L). Interestingly, the receptor binding patterns of lymphotoxin alpha and alphabeta are redundant in the human but not in the mouse system. Ligand oligomerization allowed detection of weak interactions, such as that of human TNF with mouse TNFR2. In addition, mouse APRIL exists as two different splice variants differing by a single amino acid. Although human APRIL does not interact with BAFF-R, the shorter variant of mouse APRIL exhibits weak but detectable binding to mouse BAFF-R.


Asunto(s)
Receptores del Factor de Necrosis Tumoral/metabolismo , Factores de Necrosis Tumoral/metabolismo , Empalme Alternativo , Secuencia de Aminoácidos , Animales , Membrana Celular/metabolismo , Humanos , Ligandos , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Unión Proteica , Homología de Secuencia de Aminoácido , Especificidad de la Especie
16.
Nature ; 418(6897): 562-6, 2002 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-12152085

RESUMEN

The Mre11 complex (Mre11 Rad50 Nbs1) is central to chromosomal maintenance and functions in homologous recombination, telomere maintenance and sister chromatid association. These functions all imply that the linked binding of two DNA substrates occurs, although the molecular basis for this process remains unknown. Here we present a 2.2 A crystal structure of the Rad50 coiled-coil region that reveals an unexpected dimer interface at the apex of the coiled coils in which pairs of conserved Cys-X-X-Cys motifs form interlocking hooks that bind one Zn(2+) ion. Biochemical, X-ray and electron microscopy data indicate that these hooks can join oppositely protruding Rad50 coiled-coil domains to form a flexible bridge of up to 1,200 A. This suggests a function for the long insertion in the Rad50 ABC-ATPase domain. The Rad50 hook is functional, because mutations in this motif confer radiation sensitivity in yeast and disrupt binding at the distant Mre11 nuclease interface. These data support an architectural role for the Rad50 coiled coils in forming metal-mediated bridging complexes between two DNA-binding heads. The resulting assemblies have appropriate lengths and conformational properties to link sister chromatids in homologous recombination and DNA ends in non-homologous end-joining.


Asunto(s)
Reparación del ADN , Proteínas de Unión al ADN , Endodesoxirribonucleasas/metabolismo , Exodesoxirribonucleasas/metabolismo , Proteínas Fúngicas/química , Proteínas Fúngicas/metabolismo , Recombinación Genética , Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae/química , Zinc/metabolismo , Adenosina Trifosfatasas/química , Adenosina Trifosfatasas/genética , Adenosina Trifosfatasas/metabolismo , Adenosina Trifosfatasas/ultraestructura , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Sitios de Unión , Cristalografía por Rayos X , Cisteína/genética , Cisteína/metabolismo , Dimerización , Endodesoxirribonucleasas/química , Endodesoxirribonucleasas/ultraestructura , Exodesoxirribonucleasas/química , Exodesoxirribonucleasas/ultraestructura , Proteínas Fúngicas/genética , Proteínas Fúngicas/ultraestructura , Humanos , Microscopía Electrónica , Modelos Moleculares , Datos de Secuencia Molecular , Mutación , Unión Proteica , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína , Tolerancia a Radiación/genética , Recombinación Genética/genética , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda