Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Immunol ; 206(12): 2949-2965, 2021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-34031149

RESUMEN

APCs such as myeloid dendritic cells (DCs) are key sentinels of the innate immune system. In response to pathogen recognition and innate immune stimulation, DCs transition from an immature to a mature state that is characterized by widespread changes in host gene expression, which include the upregulation of cytokines, chemokines, and costimulatory factors to protect against infection. Several transcription factors are known to drive these gene expression changes, but the mechanisms that negatively regulate DC maturation are less well understood. In this study, we identify the transcription factor IL enhancer binding factor 3 (ILF3) as a negative regulator of innate immune responses and DC maturation. Depletion of ILF3 in primary human monocyte-derived DCs led to increased expression of maturation markers and potentiated innate responses during stimulation with viral mimetics or classic innate agonists. Conversely, overexpression of short or long ILF3 isoforms (NF90 and NF110) suppressed DC maturation and innate immune responses. Through mutagenesis experiments, we found that a nuclear localization sequence in ILF3, and not its dual dsRNA-binding domains, was required for this function. Mutation of the domain associated with zinc finger motif of ILF3's NF110 isoform blocked its ability to suppress DC maturation. Moreover, RNA-sequencing analysis indicated that ILF3 regulates genes associated with cholesterol homeostasis in addition to genes associated with DC maturation. Together, our data establish ILF3 as a transcriptional regulator that restrains DC maturation and limits innate immune responses through a mechanism that may intersect with lipid metabolism.


Asunto(s)
Células Dendríticas , Transducción de Señal , Humanos , Inmunidad Innata , Monocitos , Isoformas de Proteínas/genética
2.
PLoS Pathog ; 13(11): e1006687, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-29145483

RESUMEN

Our understanding of mechanisms underlying progression from Mycobacterium tuberculosis infection to pulmonary tuberculosis disease in humans remains limited. To define such mechanisms, we followed M. tuberculosis-infected adolescents longitudinally. Blood samples from forty-four adolescents who ultimately developed tuberculosis disease ("progressors") were compared with those from 106 matched controls, who remained healthy during two years of follow up. We performed longitudinal whole blood transcriptomic analyses by RNA sequencing and plasma proteome analyses using multiplexed slow off-rate modified DNA aptamers. Tuberculosis progression was associated with sequential modulation of immunological processes. Type I/II interferon signalling and complement cascade were elevated 18 months before tuberculosis disease diagnosis, while changes in myeloid inflammation, lymphoid, monocyte and neutrophil gene modules occurred more proximally to tuberculosis disease. Analysis of gene expression in purified T cells also revealed early suppression of Th17 responses in progressors, relative to M. tuberculosis-infected controls. This was confirmed in an independent adult cohort who received BCG re-vaccination; transcript expression of interferon response genes in blood prior to BCG administration was associated with suppression of IL-17 expression by BCG-specific CD4 T cells 3 weeks post-vaccination. Our findings provide a timeline to the different immunological stages of disease progression which comprise sequential inflammatory dynamics and immune alterations that precede disease manifestations and diagnosis of tuberculosis disease. These findings have important implications for developing diagnostics, vaccination and host-directed therapies for tuberculosis. TRIAL REGISTRATION: Clincialtrials.gov, NCT01119521.


Asunto(s)
Mycobacterium tuberculosis , Linfocitos T/inmunología , Tuberculosis/microbiología , Tuberculosis/terapia , Adolescente , Niño , Progresión de la Enfermedad , Humanos , Inflamación/complicaciones , Inflamación/inmunología , Inflamación/terapia , Vacunas/uso terapéutico
3.
Lancet ; 387(10035): 2312-2322, 2016 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-27017310

RESUMEN

BACKGROUND: Identification of blood biomarkers that prospectively predict progression of Mycobacterium tuberculosis infection to tuberculosis disease might lead to interventions that combat the tuberculosis epidemic. We aimed to assess whether global gene expression measured in whole blood of healthy people allowed identification of prospective signatures of risk of active tuberculosis disease. METHODS: In this prospective cohort study, we followed up healthy, South African adolescents aged 12-18 years from the adolescent cohort study (ACS) who were infected with M tuberculosis for 2 years. We collected blood samples from study participants every 6 months and monitored the adolescents for progression to tuberculosis disease. A prospective signature of risk was derived from whole blood RNA sequencing data by comparing participants who developed active tuberculosis disease (progressors) with those who remained healthy (matched controls). After adaptation to multiplex quantitative real-time PCR (qRT-PCR), the signature was used to predict tuberculosis disease in untouched adolescent samples and in samples from independent cohorts of South African and Gambian adult progressors and controls. Participants of the independent cohorts were household contacts of adults with active pulmonary tuberculosis disease. FINDINGS: Between July 6, 2005, and April 23, 2007, we enrolled 6363 participants from the ACS study and 4466 from independent South African and Gambian cohorts. 46 progressors and 107 matched controls were identified in the ACS cohort. A 16 gene signature of risk was identified. The signature predicted tuberculosis progression with a sensitivity of 66·1% (95% CI 63·2-68·9) and a specificity of 80·6% (79·2-82·0) in the 12 months preceding tuberculosis diagnosis. The risk signature was validated in an untouched group of adolescents (p=0·018 for RNA sequencing and p=0·0095 for qRT-PCR) and in the independent South African and Gambian cohorts (p values <0·0001 by qRT-PCR) with a sensitivity of 53·7% (42·6-64·3) and a specificity of 82·8% (76·7-86) in the 12 months preceding tuberculosis. INTERPRETATION: The whole blood tuberculosis risk signature prospectively identified people at risk of developing active tuberculosis, opening the possibility for targeted intervention to prevent the disease. FUNDING: Bill & Melinda Gates Foundation, the National Institutes of Health, Aeras, the European Union, and the South African Medical Research Council.


Asunto(s)
Tuberculosis/diagnóstico , Adolescente , Adulto , Estudios de Casos y Controles , Niño , Expresión Génica , Humanos , Persona de Mediana Edad , Mycobacterium tuberculosis/genética , Estudios Prospectivos , ARN Bacteriano/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Medición de Riesgo , Factores de Riesgo , Tuberculosis/sangre , Tuberculosis/genética , Adulto Joven
4.
Mamm Genome ; 25(11-12): 549-63, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25001233

RESUMEN

Metabolic diseases such as obesity and atherosclerosis result from complex interactions between environmental factors and genetic variants. A panel of chromosome substitution strains (CSSs) was developed to characterize genetic and dietary factors contributing to metabolic diseases and other biological traits and biomedical conditions. Our goal here was to identify quantitative trait loci (QTLs) contributing to obesity, energy expenditure, and atherosclerosis. Parental strains C57BL/6 and A/J together with a panel of 21 CSSs derived from these progenitors were subjected to chronic feeding of rodent chow and atherosclerotic (females) or diabetogenic (males) test diets, and evaluated for a variety of metabolic phenotypes including several traits unique to this report, namely fat pad weights, energy balance, and atherosclerosis. A total of 297 QTLs across 35 traits were discovered, two of which provided significant protection from atherosclerosis, and several dozen QTLs modulated body weight, body composition, and circulating lipid levels in females and males. While several QTLs confirmed previous reports, most QTLs were novel. Finally, we applied the CSS quantitative genetic approach to energy balance, and identified three novel QTLs controlling energy expenditure and one QTL modulating food intake. Overall, we identified many new QTLs and phenotyped several novel traits in this mouse model of diet-induced metabolic diseases.


Asunto(s)
Aterosclerosis/genética , Metabolismo Energético/genética , Obesidad/genética , Animales , Composición Corporal , Peso Corporal , Cromosomas de los Mamíferos/genética , Dieta Alta en Grasa/efectos adversos , Femenino , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Masculino , Ratones Endogámicos C57BL , Fenotipo , Sitios de Carácter Cuantitativo
5.
J Proteome Res ; 11(5): 2653-65, 2012 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-22494190

RESUMEN

In an attempt to identify prostate cancer biomarkers with greater diagnostic and prognostic capabilities, we have developed an integrative proteomic discovery workflow focused on N-linked glycoproteins that refines the target selection process. In this work, hydrazide-based chemistry was used to identify N-linked glycopeptides from 22Rv1 prostate cancer cells cultured in vitro, which were compared with glycopeptides identified from explanted 22Rv1 murine tumor xenografts. One hundred and four human glycoproteins were identified in the former analysis and 75 in the latter, with 40 proteins overlapping between data sets. Of the 40 overlapping proteins, 80% have multiple literature references to the neoplastic process and ∼40% to prostatic neoplasms. These include a number of well-known prostate cancer-associated biomarkers, such as prostate-specific membrane antigen (PSMA). By integrating gene expression data and available literature, we identified members of the overlap data set that deserve consideration as potential prostate cancer biomarkers. Specifically, the identification of the extracellular domain of protein tyrosine phosphatase receptor type F (PTPRF) was of particular interest due to the direct involvement of PTPRF in the control of ß-catenin signaling, as well as dramatically elevated gene expression levels in the prostate compared to other tissues. In this investigation, we demonstrate that the PTPRF E-subunit is more abundant in human prostate tumor tissue compared to normal control and also detectable in murine plasma by immunoblot and ELISA. Specifically, PTPRF distinguishes between animals xenografted with the 22Rv1 cells and control animals as early as 14 days after implantation. This result suggests that the ectodomain of PTPRF has the potential to function as a novel plasma or tissue-based biomarker for prostate cancer. The workflow described adds to the literature of potential biomarker candidates for prostate cancer and demonstrates a pathway to developing new diagnostic assays.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Glicoproteínas/análisis , Neoplasias de la Próstata/diagnóstico , Proteómica/métodos , Proteínas Tirosina Fosfatasas Clase 2 Similares a Receptores/metabolismo , Animales , Biomarcadores de Tumor/sangre , Biomarcadores de Tumor/metabolismo , Western Blotting , Ensayo de Inmunoadsorción Enzimática , Glicoproteínas/genética , Glicoproteínas/metabolismo , Glicosilación , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Estructura Terciaria de Proteína , Proteínas Tirosina Fosfatasas Clase 2 Similares a Receptores/sangre , Proteínas Tirosina Fosfatasas Clase 2 Similares a Receptores/genética , Factores de Tiempo , Ensayos Antitumor por Modelo de Xenoinjerto , beta Catenina/genética , beta Catenina/metabolismo
6.
J Immunol ; 184(9): 5360-7, 2010 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-20308630

RESUMEN

Multiple TLR agonists have been shown to have antitumor effects in animal models. However, the therapeutic efficacy of TLR agonist monotherapy in cancer treatment has been limited, and the mechanisms of failure remain unknown. We demonstrate that topical treatment with a TLR-7 agonist, imiquimod, can elicit significant regression of spontaneous breast cancers in neu transgenic mice, a model of human HER-2/neu(+) breast cancer. However, tumor growth progressed once imiquimod therapy was ended. Gene expression analysis using tumor-derived RNA demonstrated that imiquimod induced high levels of IL-10 in addition to TNF-alpha and IFN-gamma. Elevated levels of circulating IL-10 were also detected in sera from imiquimod-treated mice. Elevated serum IL-10 appeared to be derived from IL-10 and dual cytokine secreting (IFN-gamma(+) and IL-10(+)) CD4(+) T cells rather than CD4(+)CD25(+)Foxp3(+) T regulatory cells, which were also induced by imiquimod treatment. Blockade of IL-10, but not TGF-beta, enhanced the antitumor effect of imiquimod by significantly prolonging survival in treated mice. These data suggest that the excessive inflammation induced by TLR agonists may result in a self-regulatory immunosuppression via IL-10 induction and that blocking IL-10 could enhance the therapeutic efficacy of these agents.


Asunto(s)
Mediadores de Inflamación/fisiología , Interleucina-10/antagonistas & inhibidores , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Neoplasias Mamarias Experimentales/patología , Glicoproteínas de Membrana/agonistas , Invasividad Neoplásica/patología , Receptor Toll-Like 7/agonistas , Enfermedad Aguda , Administración Tópica , Aminoquinolinas/metabolismo , Aminoquinolinas/uso terapéutico , Animales , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Inhibidores de Crecimiento/agonistas , Inhibidores de Crecimiento/metabolismo , Inhibidores de Crecimiento/uso terapéutico , Imiquimod , Mediadores de Inflamación/metabolismo , Mediadores de Inflamación/uso terapéutico , Interleucina-10/sangre , Ligandos , Neoplasias Mamarias Experimentales/inmunología , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/uso terapéutico , Ratones , Ratones Transgénicos , Invasividad Neoplásica/inmunología , Invasividad Neoplásica/prevención & control , Distribución Aleatoria , Receptor ErbB-2/genética , Receptor Toll-Like 7/metabolismo , Receptor Toll-Like 7/uso terapéutico , Insuficiencia del Tratamiento , Regulación hacia Arriba/inmunología
7.
Cell Rep ; 30(3): 914-931.e9, 2020 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-31968263

RESUMEN

Transcriptional programming of the innate immune response is pivotal for host protection. However, the transcriptional mechanisms that link pathogen sensing with innate activation remain poorly understood. During HIV-1 infection, human dendritic cells (DCs) can detect the virus through an innate sensing pathway, leading to antiviral interferon and DC maturation. Here, we develop an iterative experimental and computational approach to map the HIV-1 innate response circuitry in monocyte-derived DCs (MDDCs). By integrating genome-wide chromatin accessibility with expression kinetics, we infer a gene regulatory network that links 542 transcription factors with 21,862 target genes. We observe that an interferon response is required, yet insufficient, to drive MDDC maturation and identify PRDM1 and RARA as essential regulators of the interferon response and MDDC maturation, respectively. Our work provides a resource for interrogation of regulators of HIV replication and innate immunity, highlighting complexity and cooperativity in the regulatory circuit controlling the response to infection.


Asunto(s)
Células Dendríticas/metabolismo , Redes Reguladoras de Genes , VIH-1/inmunología , Inmunidad Innata/genética , Monocitos/metabolismo , Diferenciación Celular , Cromatina/metabolismo , Células Dendríticas/virología , Femenino , Regulación de la Expresión Génica , Células HEK293 , Infecciones por VIH/inmunología , Infecciones por VIH/virología , Humanos , Interferón Tipo I/metabolismo , Masculino , Monocitos/virología , Regiones Promotoras Genéticas/genética , Receptor alfa de Ácido Retinoico/metabolismo , Factores de Transcripción/metabolismo , Transcriptoma/genética
8.
Sci Immunol ; 4(37)2019 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-31350281

RESUMEN

Alveolar macrophages (AMs) are the first cells to be infected during Mycobacterium tuberculosis (M.tb.) infection. Thus, the AM response to infection is the first of many steps leading to initiation of the adaptive immune response required for efficient control of infection. A hallmark of M.tb. infection is the slow initiation of the adaptive response, yet the mechanisms responsible for this are largely unknown. To study the initial AM response to infection, we developed a system to identify, sort, and analyze M.tb.-infected AMs from the lung within the first 10 days of infection. In contrast to what has been previously described using in vitro systems, M.tb.-infected AMs up-regulate a cell-protective antioxidant transcriptional signature that is dependent on the lung environment but not bacterial virulence. Computational approaches including pathway analysis and transcription factor motif enrichment analysis identify NRF2 as a master regulator of the response. Using knockout mouse models, we demonstrate that NRF2 drives expression of the cell-protective signature in AMs and impairs the control of early bacterial growth. AMs up-regulate a substantial pro-inflammatory response to M.tb. infection only 10 days after infection, yet comparisons with bystander AMs from the same infected animals demonstrate that M.tb.-infected AMs generate a less robust inflammatory response than the uninfected cells around them. Our findings demonstrate that the initial macrophage response to M.tb. in the lung is far less inflammatory than has previously been described by in vitro systems and may impede the overall host response to infection.


Asunto(s)
Macrófagos Alveolares/inmunología , Macrófagos Alveolares/metabolismo , Mycobacterium tuberculosis/inmunología , Factor 2 Relacionado con NF-E2/metabolismo , Transcripción Genética , Tuberculosis Pulmonar/genética , Tuberculosis Pulmonar/inmunología , Animales , Femenino , Macrófagos Alveolares/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Tuberculosis Pulmonar/microbiología , Tuberculosis Pulmonar/patología
9.
Cell Host Microbe ; 23(3): 366-381.e9, 2018 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-29544097

RESUMEN

Myeloid dendritic cells (DCs) have the innate capacity to sense pathogens and orchestrate immune responses. However, DCs do not mount efficient immune responses to HIV-1, primarily due to restriction of virus reverse transcription, which prevents accumulation of viral cDNA and limits its detection through the cGAS-STING pathway. By allowing reverse transcription to proceed, we find that DCs detect HIV-1 in distinct phases, before and after virus integration. Blocking integration suppresses, but does not abolish, activation of the transcription factor IRF3, downstream interferon (IFN) responses, and DC maturation. Consistent with two stages of detection, HIV-1 "primes" chromatin accessibility of innate immune genes before and after integration. Once primed, robust IFN responses can be unmasked by agonists of the innate adaptor protein, MyD88, through a process that requires cGAS, STING, IRF3, and nuclear factor κB. Thus, HIV-1 replication increases material available for sensing, and discrete inflammatory inputs tune cGAS signaling to drive DC maturation.


Asunto(s)
Cromatina/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , VIH-1/inmunología , Interacciones Huésped-Patógeno/inmunología , Interferones/metabolismo , Línea Celular , Femenino , Células HEK293 , Infecciones por VIH/metabolismo , VIH-1/patogenicidad , Humanos , Inmunidad Innata/genética , Inmunidad Innata/inmunología , Factor 3 Regulador del Interferón/metabolismo , Interferón Tipo I/metabolismo , Masculino , Proteínas de la Membrana/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , FN-kappa B/metabolismo , Nucleotidiltransferasas/metabolismo , Transcripción Reversa , Transducción de Señal , Células THP-1 , Integración Viral , Replicación Viral
10.
Nat Med ; 24(2): 130-143, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29334373

RESUMEN

Despite widespread use of the bacille Calmette-Guérin (BCG) vaccine, tuberculosis (TB) remains a leading cause of global mortality from a single infectious agent (Mycobacterium tuberculosis or Mtb). Here, over two independent Mtb challenge studies, we demonstrate that subcutaneous vaccination of rhesus macaques (RMs) with rhesus cytomegalovirus vectors encoding Mtb antigen inserts (hereafter referred to as RhCMV/TB)-which elicit and maintain highly effector-differentiated, circulating and tissue-resident Mtb-specific CD4+ and CD8+ memory T cell responses-can reduce the overall (pulmonary and extrapulmonary) extent of Mtb infection and disease by 68%, as compared to that in unvaccinated controls, after intrabronchial challenge with the Erdman strain of Mtb at ∼1 year after the first vaccination. Fourteen of 34 RhCMV/TB-vaccinated RMs (41%) across both studies showed no TB disease by computed tomography scans or at necropsy after challenge (as compared to 0 of 17 unvaccinated controls), and ten of these RMs were Mtb-culture-negative for all tissues, an exceptional long-term vaccine effect in the RM challenge model with the Erdman strain of Mtb. These results suggest that complete vaccine-mediated immune control of highly pathogenic Mtb is possible if immune effector responses can intercept Mtb infection at its earliest stages.


Asunto(s)
Mycobacterium tuberculosis/inmunología , Tuberculosis/inmunología , Animales , Vacuna BCG/inmunología , Citomegalovirus/inmunología , Macaca mulatta/inmunología
11.
EBioMedicine ; 7: 278-86, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27322481

RESUMEN

BACKGROUND: We report a first-in-human trial evaluating safety and immunogenicity of a recombinant BCG, AERAS-422, over-expressing TB antigens Ag85A, Ag85B, and Rv3407 and expressing mutant perfringolysin. METHODS: This was a randomized, double-blind, dose-escalation trial in HIV-negative, healthy adult, BCG-naïve volunteers, negative for prior exposure to Mtb, at one US clinical site. Volunteers were randomized 2:1 at each dose level to receive a single intradermal dose of AERAS-422 (>10(5)-<10(6)CFU=low dose, ≥10(6)-<10(7)CFU=high dose) or non-recombinant Tice BCG (1-8×10(5)CFU). Randomization used an independently prepared randomly generated sequence of treatment assignments. The primary and secondary outcomes were safety and immunogenicity, respectively, assessed in all participants through 182days post-vaccination. ClinicalTrials.gov registration number: NCT01340820. FINDINGS: Between Nov 2010 and Aug 2011, 24 volunteers were enrolled (AERAS-422 high dose, n=8; AERAS-422 low dose, n=8; Tice BCG, n=8); all were included in the safety and immunogenicity analyses. All 24 subjects had at least one adverse event, primarily expected local reactions. High dose AERAS-422 vaccination induced Ag85A- and Ag85B-specific lymphoproliferative responses and marked anti-mycobacterial activity in a whole blood bactericidal activity culture assay (WBA), but was associated with varicella zoster virus (VZV) reactivation in two vaccinees. These volunteers displayed high BCG-specific IFN-γ responses pre- and post-vaccination possibly predisposing them to autocrine/paracrine negative regulation of immune control of latent VZV. A systems biology transcriptomal approach identified positive correlations between post-vaccination T cell expression modules and WBA, and negative correlations between post-vaccination monocyte expression modules and WBA. The expression of one key macrophage marker (F4/80) was constitutively elevated in the two volunteers with zoster. INTERPRETATION: The unexpected development of VZV in two of eight healthy adult vaccine recipients resulted in discontinuation of AERAS-422 vaccine development. Immunological and transcriptomal data identified correlations with the development of TB immunity and VZV that require further investigation. FUNDING: Aeras, FDA, Bill and Melinda Gates Foundation.


Asunto(s)
Vacuna BCG/administración & dosificación , Vacuna BCG/inmunología , Herpesvirus Humano 3/fisiología , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/inmunología , Aciltransferasas/inmunología , Aciltransferasas/metabolismo , Adulto , Antígenos Bacterianos/inmunología , Antígenos Bacterianos/metabolismo , Vacuna BCG/efectos adversos , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/metabolismo , Toxinas Bacterianas/inmunología , Toxinas Bacterianas/metabolismo , Relación Dosis-Respuesta a Droga , Voluntarios Sanos , Proteínas Hemolisinas/inmunología , Proteínas Hemolisinas/metabolismo , Humanos , Masculino , Vacunas Sintéticas/efectos adversos , Activación Viral , Adulto Joven
12.
Cancer Res ; 73(5): 1502-13, 2013 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-23269276

RESUMEN

We assessed the autoantibody repertoire of a mouse model engineered to develop breast cancer and the repertoire of autoantibodies in human plasmas collected at a preclinical time point and at the time of clinical diagnosis of breast cancer. In seeking to identify common pathways, networks, and protein families associated with the humoral response, we elucidated the dynamic nature of tumor antigens and autoantibody interactions. Lysate proteins from an immortalized cell line from a MMTV-neu mouse model and from MCF7 human breast cancers were spotted onto nitrocellulose microarrays and hybridized with mouse and human plasma samples, respectively. Immunoglobulin-based plasma immunoreactivity against glycolysis and spliceosome proteins was a predominant feature observed both in tumor-bearing mice and in prediagnostic human samples. Interestingly, autoantibody reactivity was more pronounced further away than closer to diagnosis. We provide evidence for dynamic changes in autoantibody reactivity with tumor development and progression that may depend, in part, on the extent of antigen-antibody interactions.


Asunto(s)
Autoanticuerpos/sangre , Neoplasias de la Mama/inmunología , Glucólisis/inmunología , Empalmosomas/inmunología , Anciano , Animales , Anticuerpos Antineoplásicos/sangre , Reacciones Antígeno-Anticuerpo , Antígenos de Neoplasias/inmunología , Neoplasias de la Mama/diagnóstico , Línea Celular Tumoral , Femenino , Humanos , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Posmenopausia , Empalmosomas/metabolismo , Factores de Tiempo
13.
Cancer Res ; 72(8): 1935-42, 2012 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-22367215

RESUMEN

Although the identification of peripheral blood biomarkers would enhance early detection strategies for breast cancer, the discovery of protein markers has been challenging. In this study, we sought to identify coordinated changes in plasma proteins associated with breast cancer based on large-scale quantitative mass spectrometry. We analyzed plasma samples collected up to 74 weeks before diagnosis from 420 estrogen receptor (ER)(+) cases and matched controls enrolled in the Women's Health Initiative cohort. A gene set enrichment analysis was applied to 467 quantified proteins, linking their corresponding genes to particular biologic pathways. On the basis of differences in the concentration of individual proteins, glycolysis pathway proteins exhibited a statistically significant difference between cases and controls. In particular, the enrichment was observed among cases in which blood was drawn closer to diagnosis (effect size for the 0-38 weeks prediagnostic group, 1.91; P, 8.3E-05). Analysis of plasmas collected at the time of diagnosis from an independent set of cases and controls confirmed upregulated levels of glycolysis proteins among cases relative to controls. Together, our findings indicate that the concomitant release of glycolysis proteins into the plasma is a pathophysiologic event that precedes a diagnosis of ER(+) breast cancer.


Asunto(s)
Biomarcadores de Tumor/sangre , Neoplasias de la Mama/sangre , Neoplasias de la Mama/diagnóstico , Glucólisis/fisiología , Anciano , Neoplasias de la Mama/genética , Estudios de Casos y Controles , Detección Precoz del Cáncer/métodos , Femenino , Humanos , Espectrometría de Masas , Persona de Mediana Edad , Proteómica/métodos , Receptores de Estrógenos/genética
14.
Nat Biotechnol ; 29(7): 625-34, 2011 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-21685906

RESUMEN

High-throughput technologies can now identify hundreds of candidate protein biomarkers for any disease with relative ease. However, because there are no assays for the majority of proteins and de novo immunoassay development is prohibitively expensive, few candidate biomarkers are tested in clinical studies. We tested whether the analytical performance of a biomarker identification pipeline based on targeted mass spectrometry would be sufficient for data-dependent prioritization of candidate biomarkers, de novo development of assays and multiplexed biomarker verification. We used a data-dependent triage process to prioritize a subset of putative plasma biomarkers from >1,000 candidates previously identified using a mouse model of breast cancer. Eighty-eight novel quantitative assays based on selected reaction monitoring mass spectrometry were developed, multiplexed and evaluated in 80 plasma samples. Thirty-six proteins were verified as being elevated in the plasma of tumor-bearing animals. The analytical performance of this pipeline suggests that it should support the use of an analogous approach with human samples.


Asunto(s)
Biomarcadores de Tumor/sangre , Análisis Químico de la Sangre/métodos , Espectrometría de Masas/métodos , Proteínas de Neoplasias/sangre , Neoplasias Experimentales/sangre , Mapeo Peptídico/métodos , Proteoma/análisis , Animales , Ratones , Proteómica/métodos
15.
Proteomics Clin Appl ; 5(3-4): 179-88, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21448875

RESUMEN

PURPOSE: We generated extensive transcriptional and proteomic profiles from a Her2-driven mouse model of breast cancer that closely recapitulates human breast cancer. This report makes these data publicly available in raw and processed forms, as a resource to the community. Importantly, we previously made biospecimens from this same mouse model freely available through a sample repository, so researchers can obtain samples to test biological hypotheses without the need of breeding animals and collecting biospecimens. EXPERIMENTAL DESIGN: Twelve datasets are available, encompassing 841 LC-MS/MS experiments (plasma and tissues) and 255 microarray analyses of multiple tissues (thymus, spleen, liver, blood cells, and breast). Cases and controls were rigorously paired to avoid bias. RESULTS: In total, 18,880 unique peptides were identified (PeptideProphet peptide error rate ≤1%), with 3884 and 1659 non-redundant protein groups identified in plasma and tissue datasets, respectively. Sixty-one of these protein groups overlapped between cancer plasma and cancer tissue. CONCLUSIONS AND CLINICAL RELEVANCE: These data are of use for advancing our understanding of cancer biology, for software and quality control tool development, investigations of analytical variation in MS/MS data, and selection of proteotypic peptides for multiple reaction monitoring-MS. The availability of these datasets will contribute positively to clinical proteomics.


Asunto(s)
Neoplasias de la Mama/genética , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Proteoma/análisis , Proteoma/genética , Receptor ErbB-2/genética , Transcripción Genética/genética , Animales , Bases de Datos de Proteínas , Ratones , Ratones Transgénicos , Proteómica , Receptor ErbB-2/análisis , Espectrometría de Masas en Tándem
16.
PLoS One ; 5(6): e11137, 2010 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-20559444

RESUMEN

BACKGROUND: We used intensive modern proteomics approaches to identify predictive proteins in ovary cancer. We identify up-regulated proteins in both serum and peritoneal fluid. To evaluate the overall performance of the approach we track the behavior of 20 validated markers across these experiments. METHODOLOGY: Mass spectrometry based quantitative proteomics following extensive protein fractionation was used to compare serum of women with serous ovarian cancer to healthy women and women with benign ovarian tumors. Quantitation was achieved by isotopically labeling cysteine amino acids. Label-free mass spectrometry was used to compare peritoneal fluid taken from women with serous ovarian cancer and those with benign tumors. All data were integrated and annotated based on whether the proteins have been previously validated using antibody-based assays. FINDINGS: We selected 54 quantified serum proteins and 358 peritoneal fluid proteins whose case-control differences exceeded a predefined threshold. Seventeen proteins were quantified in both materials and 14 are extracellular. Of 19 validated markers that were identified all were found in cancer peritoneal fluid and a subset of 7 were quantified in serum, with one of these proteins, IGFBP1, newly validated here. CONCLUSION: Proteome profiling applied to symptomatic ovarian cancer cases identifies a large number of up-regulated serum proteins, many of which are or have been confirmed by immunoassays. The number of currently known validated markers is highest in peritoneal fluid, but they make up a higher percentage of the proteins observed in both serum and peritoneal fluid, suggesting that the 10 additional markers in this group may be high quality candidates.


Asunto(s)
Líquido Ascítico/metabolismo , Proteínas Sanguíneas/metabolismo , Neoplasias Ováricas/sangre , Proteómica , Regulación hacia Arriba , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Proteína 1 de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Espectrometría de Masas , Neoplasias Ováricas/metabolismo
17.
Genome Med ; 2(7): 48, 2010 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-20667078

RESUMEN

BACKGROUND: Coronary heart disease (CHD) and stroke were key outcomes in the Women's Health Initiative (WHI) randomized trials of postmenopausal estrogen and estrogen plus progestin therapy. We recently reported a large number of changes in blood protein concentrations in the first year following randomization in these trials using an in-depth quantitative proteomics approach. However, even though many affected proteins are in pathways relevant to the observed clinical effects, the relationships of these proteins to CHD and stroke risk among postmenopausal women remains substantially unknown. METHODS: The same in-depth proteomics platform was applied to plasma samples, obtained at enrollment in the WHI Observational Study, from 800 women who developed CHD and 800 women who developed stroke during cohort follow-up, and from 1-1 matched controls. A plasma pooling strategy, followed by extensive fractionation prior to mass spectrometry, was used to identify proteins related to disease incidence, and the overlap of these proteins with those affected by hormone therapy was examined. Replication studies, using enzyme-linked-immunosorbent assay (ELISA), were carried out in the WHI hormone therapy trial cohorts. RESULTS: Case versus control concentration differences were suggested for 37 proteins (nominal P < 0.05) for CHD, with three proteins, beta-2 microglobulin (B2M), alpha-1-acid glycoprotein 1 (ORM1), and insulin-like growth factor binding protein acid labile subunit (IGFALS) having a false discovery rate < 0.05. Corresponding numbers for stroke were 47 proteins with nominal P < 0.05, three of which, apolipoprotein A-II precursor (APOA2), peptidyl-prolyl isomerase A (PPIA), and insulin-like growth factor binding protein 4 (IGFBP4), have a false discovery rate < 0.05. Other proteins involved in insulin-like growth factor signaling were also highly ranked. The associations of B2M with CHD (P < 0.001) and IGFBP4 with stroke (P = 0.005) were confirmed using ELISA in replication studies, and changes in these proteins following the initiation of hormone therapy use were shown to have potential to help explain hormone therapy effects on those diseases. CONCLUSIONS: In-depth proteomic discovery analysis of prediagnostic plasma samples identified B2M and IGFBP4 as risk markers for CHD and stroke respectively, and provided a number of candidate markers of disease risk and candidate mediators of hormone therapy effects on CHD and stroke. CLINICAL TRIALS REGISTRATION: ClinicalTrials.gov identifier: NCT00000611.

18.
Cancer Res ; 70(21): 8598-606, 2010 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-20959476

RESUMEN

Applying advanced proteomic technologies to prospectively collected specimens from large studies is one means of identifying preclinical changes in plasma proteins that are potentially relevant to the early detection of diseases such as breast cancer. We conducted 14 independent quantitative proteomics experiments comparing pooled plasma samples collected from 420 estrogen receptor-positive (ER(+)) breast cancer patients ≤17 months before their diagnosis and matched controls. Based on the more than 3.4 million tandem mass spectra collected in the discovery set, 503 proteins were quantified, of which 57 differentiated cases from controls with a P value of <0.1. Seven of these proteins, for which quantitative ELISA assays were available, were assessed in an independent validation set. Of these candidates, epidermal growth factor receptor (EGFR) was validated as a predictor of breast cancer risk in an independent set of preclinical plasma samples for women overall [odds ratio (OR), 1.44; P = 0.0008] and particularly for current users of estrogen plus progestin (E + P) menopausal hormone therapy (OR, 2.49; P = 0.0001). Among current E + P users, the EGFR sensitivity for breast cancer risk was 31% with 90% specificity. Whereas the sensitivity and specificity of EGFR are insufficient for a clinically useful early detection biomarker, this study suggests that proteins that are elevated preclinically in women who go on to develop breast cancer can be discovered and validated using current proteomic technologies. Further studies are warranted to examine the role of EGFR and to discover and validate other proteins that could potentially be used for early detection of breast cancer.


Asunto(s)
Neoplasias de la Mama/diagnóstico , Carcinoma Ductal de Mama/diagnóstico , Carcinoma Lobular/diagnóstico , Receptores ErbB/sangre , Terapia de Reemplazo de Hormonas , Secuencia de Aminoácidos , Mama/metabolismo , Mama/patología , Neoplasias de la Mama/sangre , Neoplasias de la Mama/tratamiento farmacológico , Carcinoma Ductal de Mama/sangre , Carcinoma Ductal de Mama/tratamiento farmacológico , Carcinoma Lobular/sangre , Carcinoma Lobular/tratamiento farmacológico , Estudios de Casos y Controles , Cromatografía Liquida , Estudios de Cohortes , Quimioterapia Combinada , Electroforesis en Gel Bidimensional , Estrógenos/uso terapéutico , Femenino , Estudios de Seguimiento , Humanos , Menopausia , Datos de Secuencia Molecular , Oportunidad Relativa , Progestinas/uso terapéutico , Pronóstico , Estudios Prospectivos , Proteómica , Receptores de Estrógenos/metabolismo , Sensibilidad y Especificidad , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Tasa de Supervivencia , Espectrometría de Masas en Tándem
19.
Genome Med ; 1(12): 121, 2009 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-20034393

RESUMEN

BACKGROUND: Women's Health Initiative randomized trials of postmenopausal hormone therapy reported intervention effects on several clinical outcomes, with some important differences between estrogen alone and estrogen plus progestin. The biologic mechanisms underlying these effects, and these differences, have yet to be fully elucidated. METHODS: Baseline serum samples were compared with samples drawn 1 year later for 50 women assigned to active hormone therapy in both the estrogen-plus-progestin and estrogen-alone randomized trials, by applying an in-depth proteomic discovery platform to serum pools from 10 women per pool. RESULTS: In total, 378 proteins were quantified in two or more of the 10 pooled serum comparisons, by using strict identification criteria. Of these, 169 (44.7%) showed evidence (nominal P < 0.05) of change in concentration between baseline and 1 year for one or both of estrogen-plus-progestin and estrogen-alone groups. Quantitative changes were highly correlated between the two hormone-therapy preparations. A total of 98 proteins had false discovery rates < 0.05 for change with estrogen plus progestin, compared with 94 for estrogen alone. Of these, 84 had false discovery rates <0.05 for both preparations. The observed changes included multiple proteins relevant to coagulation, inflammation, immune response, metabolism, cell adhesion, growth factors, and osteogenesis. Evidence of differential changes also was noted between the hormone preparations, with the strongest evidence in growth factor and inflammation pathways. CONCLUSIONS: Serum proteomic analyses yielded a large number of proteins similarly affected by estrogen plus progestin and by estrogen alone and identified some proteins and pathways that appear to be differentially affected between the two hormone preparations; this may explain their distinct clinical effects.

20.
Arthritis Rheum ; 58(5): 1465-74, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18438864

RESUMEN

OBJECTIVE: We attempted to elucidate possible pathogenetic mechanisms in scleroderma by analysis of gene expression patterns of purified monocytes and lymphocytes, as well as protein profiles of cytokines and growth factors. METHODS: Expression analysis was performed on messenger RNA (mRNA) from cells that had been purified with magnetic beads. Plasma samples from the same patients were used for multiplex cytokine analysis. Potential sources of proteins were also examined by in situ hybridization of skin specimens. RESULTS: A total of 1,800 genes from monocytes and 863 genes from CD4+ T cells were differentially expressed in scleroderma patients. As observed by other investigators using unfractionated peripheral blood cells from patients with autoimmune connective tissue diseases, the cell type-specific analyses of our scleroderma samples showed expression of genes suggesting the presence of interferon-alpha (IFNalpha), despite the apparent absence of this cytokine in plasma. IFNalpha RNA was, however, expressed at enhanced levels in vascular and perivascular cells in scleroderma skin samples. While levels of interleukin-1alpha (IL-1alpha) and IL-16 were among 10 proteins found to be significantly elevated in scleroderma patients, none of the large panel of plasma cytokines we analyzed correlated with the expression levels of putative IFN response genes. CONCLUSION: The pattern of up-regulation of mRNA in both the monocytes and CD4 lymphocytes of scleroderma patients, together with the detection of IFNalpha RNA in the microvasculature, suggests that leukocytes respond to this cytokine locally in the vessels. Detection of high levels of IL-1alpha and IL-16 in plasma and the independence of these protein levels from the IFN signature, implicates an independent contribution of other cytokines to immune activation and/or inflammation in scleroderma.


Asunto(s)
Linfocitos/metabolismo , Monocitos/metabolismo , ARN Mensajero/biosíntesis , Esclerodermia Difusa/sangre , Esclerodermia Limitada/sangre , Adulto , Anciano , Proteínas Sanguíneas/análisis , Femenino , Humanos , Persona de Mediana Edad , Esclerodermia Difusa/genética , Esclerodermia Limitada/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA