Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 184(15): 3915-3935.e21, 2021 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-34174187

RESUMEN

Emerging evidence indicates a fundamental role for the epigenome in immunity. Here, we mapped the epigenomic and transcriptional landscape of immunity to influenza vaccination in humans at the single-cell level. Vaccination against seasonal influenza induced persistently diminished H3K27ac in monocytes and myeloid dendritic cells (mDCs), which was associated with impaired cytokine responses to Toll-like receptor stimulation. Single-cell ATAC-seq analysis revealed an epigenomically distinct subcluster of monocytes with reduced chromatin accessibility at AP-1-targeted loci after vaccination. Similar effects were observed in response to vaccination with the AS03-adjuvanted H5N1 pandemic influenza vaccine. However, this vaccine also stimulated persistently increased chromatin accessibility at interferon response factor (IRF) loci in monocytes and mDCs. This was associated with elevated expression of antiviral genes and heightened resistance to the unrelated Zika and Dengue viruses. These results demonstrate that vaccination stimulates persistent epigenomic remodeling of the innate immune system and reveal AS03's potential as an epigenetic adjuvant.


Asunto(s)
Epigenómica , Inmunidad/genética , Vacunas contra la Influenza/genética , Vacunas contra la Influenza/inmunología , Análisis de la Célula Individual , Transcripción Genética , Vacunación , Adolescente , Adulto , Antibacterianos/farmacología , Antígenos CD34/metabolismo , Antivirales/farmacología , Reprogramación Celular , Cromatina/metabolismo , Citocinas/biosíntesis , Combinación de Medicamentos , Femenino , Regulación de la Expresión Génica , Histonas/metabolismo , Humanos , Inmunidad Innata/genética , Subtipo H5N1 del Virus de la Influenza A/efectos de los fármacos , Subtipo H5N1 del Virus de la Influenza A/inmunología , Interferón Tipo I/metabolismo , Masculino , Células Mieloides/metabolismo , Polisorbatos/farmacología , Escualeno/farmacología , Receptores Toll-Like/metabolismo , Factor de Transcripción AP-1/metabolismo , Transcriptoma/genética , Adulto Joven , alfa-Tocoferol/farmacología
2.
Nat Immunol ; 22(6): 711-722, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34017121

RESUMEN

Chromatin undergoes extensive reprogramming during immune cell differentiation. Here we report the repression of controlled histone H3 amino terminus proteolytic cleavage (H3ΔN) during monocyte-to-macrophage development. This abundant histone mark in human peripheral blood monocytes is catalyzed by neutrophil serine proteases (NSPs) cathepsin G, neutrophil elastase and proteinase 3. NSPs are repressed as monocytes mature into macrophages. Integrative epigenomic analysis reveals widespread H3ΔN distribution across the genome in a monocytic cell line and primary monocytes, which becomes largely undetectable in fully differentiated macrophages. H3ΔN is enriched at permissive chromatin and actively transcribed genes. Simultaneous NSP depletion in monocytic cells results in H3ΔN loss and further increase in chromatin accessibility, which likely primes the chromatin for gene expression reprogramming. Importantly, H3ΔN is reduced in monocytes from patients with systemic juvenile idiopathic arthritis, an autoinflammatory disease with prominent macrophage involvement. Overall, we uncover an epigenetic mechanism that primes the chromatin to facilitate macrophage development.


Asunto(s)
Artritis Juvenil/inmunología , Diferenciación Celular/inmunología , Epigénesis Genética/inmunología , Histonas/metabolismo , Leucocitos Mononucleares/metabolismo , Macrófagos/inmunología , Adolescente , Artritis Juvenil/sangre , Artritis Juvenil/genética , Sistemas CRISPR-Cas/genética , Catepsina G/genética , Catepsina G/metabolismo , Diferenciación Celular/genética , Núcleo Celular/metabolismo , Niño , Preescolar , Cromatina/metabolismo , Pruebas de Enzimas , Epigenómica , Femenino , Técnicas de Inactivación de Genes , Humanos , Células Jurkat , Elastasa de Leucocito/genética , Elastasa de Leucocito/metabolismo , Leucocitos Mononucleares/inmunología , Macrófagos/metabolismo , Masculino , Mieloblastina/genética , Mieloblastina/metabolismo , Cultivo Primario de Células , Proteolisis , RNA-Seq , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Células THP-1 , Adulto Joven
3.
Cell ; 173(6): 1385-1397.e14, 2018 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-29706550

RESUMEN

Post-translational modifications of histone proteins and exchanges of histone variants of chromatin are central to the regulation of nearly all DNA-templated biological processes. However, the degree and variability of chromatin modifications in specific human immune cells remain largely unknown. Here, we employ a highly multiplexed mass cytometry analysis to profile the global levels of a broad array of chromatin modifications in primary human immune cells at the single-cell level. Our data reveal markedly different cell-type- and hematopoietic-lineage-specific chromatin modification patterns. Differential analysis between younger and older adults shows that aging is associated with increased heterogeneity between individuals and elevated cell-to-cell variability in chromatin modifications. Analysis of a twin cohort unveils heritability of chromatin modifications and demonstrates that aging-related chromatin alterations are predominantly driven by non-heritable influences. Together, we present a powerful platform for chromatin and immunology research. Our discoveries highlight the profound impacts of aging on chromatin modifications.


Asunto(s)
Envejecimiento , Cromatina/química , Epigénesis Genética , Adolescente , Adulto , Anciano , Linaje de la Célula , Separación Celular , Enfermedades en Gemelos , Femenino , Citometría de Flujo , Histonas/metabolismo , Humanos , Sistema Inmunológico , Inmunofenotipificación , Leucocitos Mononucleares/citología , Masculino , Persona de Mediana Edad , Monocitos/citología , Análisis de Componente Principal , Procesamiento Proteico-Postraduccional , Sistema de Registros , Adulto Joven
4.
Nat Immunol ; 12(1): 29-36, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21131967

RESUMEN

Signaling via the methylation of lysine residues in proteins has been linked to diverse biological and disease processes, yet the catalytic activity and substrate specificity of many human protein lysine methyltransferases (PKMTs) are unknown. We screened over 40 candidate PKMTs and identified SETD6 as a methyltransferase that monomethylated chromatin-associated transcription factor NF-κB subunit RelA at Lys310 (RelAK310me1). SETD6-mediated methylation rendered RelA inert and attenuated RelA-driven transcriptional programs, including inflammatory responses in primary immune cells. RelAK310me1 was recognized by the ankryin repeat of the histone methyltransferase GLP, which under basal conditions promoted a repressed chromatin state at RelA target genes through GLP-mediated methylation of histone H3 Lys9 (H3K9). NF-κB-activation-linked phosphorylation of RelA at Ser311 by protein kinase C-ζ (PKC-ζ) blocked the binding of GLP to RelAK310me1 and relieved repression of the target gene. Our findings establish a previously uncharacterized mechanism by which chromatin signaling regulates inflammation programs.


Asunto(s)
Artritis Reumatoide/inmunología , FN-kappa B/metabolismo , Proteína Metiltransferasas/metabolismo , Factor de Transcripción ReIA/metabolismo , Artritis Reumatoide/genética , Artritis Reumatoide/metabolismo , Ensamble y Desensamble de Cromatina/genética , Metilación de ADN , Células HEK293 , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Humanos , Inflamación , Lisina/metabolismo , FN-kappa B/genética , FN-kappa B/inmunología , Unión Proteica/genética , Proteína Metiltransferasas/genética , Proteína Metiltransferasas/inmunología , ARN Interferente Pequeño/genética , Transducción de Señal/genética , Transducción de Señal/inmunología , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIA/inmunología
5.
Genes Dev ; 28(16): 1758-71, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-25128496

RESUMEN

The dynamic reversible methylation of lysine residues on histone proteins is central to chromatin biology. Key components are demethylase enzymes, which remove methyl moieties from lysine residues. KDM2A, a member of the Jumonji C domain-containing histone lysine demethylase family, specifically targets lower methylation states of H3K36. Here, structural studies reveal that H3K36 specificity for KDM2A is mediated by the U-shaped threading of the H3K36 peptide through a catalytic groove within KDM2A. The side chain of methylated K36 inserts into the catalytic pocket occupied by Ni(2+) and cofactor, where it is positioned and oriented for demethylation. Key residues contributing to K36me specificity on histone H3 are G33 and G34 (positioned within a narrow channel), P38 (a turn residue), and Y41 (inserts into its own pocket). Given that KDM2A was found to also bind the H3K36me3 peptide, we postulate that steric constraints could prevent α-ketoglutarate from undergoing an "off-line"-to-"in-line" transition necessary for the demethylation reaction. Furthermore, structure-guided substitutions of residues in the KDM2A catalytic pocket abrogate KDM2A-mediated functions important for suppression of cancer cell phenotypes. Together, our results deduce insights into the molecular basis underlying KDM2A regulation of the biologically important methylated H3K36 mark.


Asunto(s)
Histonas/metabolismo , Histona Demetilasas con Dominio de Jumonji/química , Histona Demetilasas con Dominio de Jumonji/metabolismo , Modelos Moleculares , Animales , Sitios de Unión , Línea Celular , Inestabilidad Genómica , Histona Demetilasas con Dominio de Jumonji/genética , Metilación , Ratones , Mutación , Unión Proteica , Estructura Cuaternaria de Proteína
6.
Mol Cell ; 44(4): 609-20, 2011 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-22099308

RESUMEN

The histone lysine methyltransferase NSD2 (MMSET/WHSC1) is implicated in diverse diseases and commonly overexpressed in multiple myeloma due to a recurrent t(4;14) chromosomal translocation. However, the precise catalytic activity of NSD2 is obscure, preventing progress in understanding how this enzyme influences chromatin biology and myeloma pathogenesis. Here, we show that dimethylation of histone H3 at lysine 36 (H3K36me2) is the principal chromatin-regulatory activity of NSD2. Catalysis of H3K36me2 by NSD2 is sufficient for gene activation. In t(4;14)-positive myeloma cells, the normal genome-wide and gene-specific distribution of H3K36me2 is obliterated, creating a chromatin landscape that selects for a transcription profile favorable for myelomagenesis. Catalytically active NSD2 confers xenograft tumor formation upon t(4;14)-negative cells and promotes oncogenic transformation of primary cells in an H3K36me2-dependent manner. Together, our findings establish H3K36me2 as the primary product generated by NSD2 and demonstrate that genomic disorganization of this canonical chromatin mark by NSD2 initiates oncogenic programming.


Asunto(s)
Transformación Celular Neoplásica , Regulación de la Expresión Génica , N-Metiltransferasa de Histona-Lisina , Histonas/metabolismo , Lisina/metabolismo , Mieloma Múltiple/genética , Proteínas Recombinantes/metabolismo , Proteínas Represoras , Transducción de Señal/genética , Animales , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Cromatina , Perfilación de la Expresión Génica , Estudio de Asociación del Genoma Completo , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/genética , Humanos , Metilación , Ratones , Ratones SCID , Mieloma Múltiple/enzimología , Mieloma Múltiple/patología , Proteínas Recombinantes/genética , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Transcripción Genética , Translocación Genética , Ensayos Antitumor por Modelo de Xenoinjerto
7.
J Med Internet Res ; 21(6): e12394, 2019 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-31162126

RESUMEN

BACKGROUND: Social media technology such as Twitter allows users to share their thoughts, feelings, and opinions online. The growing body of social media data is becoming a central part of infodemiology research as these data can be combined with other public health datasets (eg, physical activity levels) to provide real-time monitoring of psychological and behavior outcomes that inform health behaviors. Currently, it is unclear whether Twitter data can be used to monitor physical activity levels. OBJECTIVE: The aim of this study was to establish the feasibility of using Twitter data to monitor physical activity levels by assessing whether the frequency and sentiment of physical activity-related tweets were associated with physical activity levels across the United States. METHODS: Tweets were collected from Twitter's application programming interface (API) between January 10, 2017 and January 2, 2018. We used Twitter's garden hose method of collecting tweets, which provided a random sample of approximately 1% of all tweets with location metadata falling within the United States. Geotagged tweets were filtered. A list of physical activity-related hashtags was collected and used to further classify these geolocated tweets. Twitter data were merged with physical activity data collected as part of the Behavioral Risk Factor Surveillance System. Multiple linear regression models were fit to assess the relationship between physical activity-related tweets and physical activity levels by county while controlling for population and socioeconomic status measures. RESULTS: During the study period, 442,959,789 unique tweets were collected, of which 64,005,336 (14.44%) were geotagged with latitude and longitude coordinates. Aggregated data were obtained for a total of 3138 counties in the United States. The mean county-level percentage of physically active individuals was 74.05% (SD 5.2) and 75.30% (SD 4.96) after adjusting for age. The model showed that the percentage of physical activity-related tweets was significantly associated with physical activity levels (beta=.11; SE 0.2; P<.001) and age-adjusted physical activity (beta=.10; SE 0.20; P<.001) on a county level while adjusting for both Gini index and education level. However, the overall explained variance of the model was low (R2=.11). The sentiment of the physical activity-related tweets was not a significant predictor of physical activity level and age-adjusted physical activity on a county level after including the Gini index and education level in the model (P>.05). CONCLUSIONS: Social media data may be a valuable tool for public health organizations to monitor physical activity levels, as it can overcome the time lag in the reporting of physical activity epidemiology data faced by traditional research methods (eg, surveys and observational studies). Consequently, this tool may have the potential to help public health organizations better mobilize and target physical activity interventions.


Asunto(s)
Ejercicio Físico/psicología , Medios de Comunicación Sociales/normas , Humanos
8.
Clin Immunol ; 196: 40-48, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29960011

RESUMEN

Modifications of histone proteins are fundamental to the regulation of epigenetic phenotypes. Dysregulations of histone modifications have been linked to the pathogenesis of diverse human diseases. However, identifying differential histone modifications in patients with immune-mediated diseases has been challenging, in part due to the lack of a powerful analytic platform to study histone modifications in the complex human immune system. We recently developed a highly multiplexed platform, Epigenetic landscape profiling using cytometry by Time-Of-Flight (EpiTOF), to analyze the global levels of a broad array of histone modifications in single cells using mass cytometry. In this review, we summarize the development of EpiTOF and discuss its potential applications in biomedical research. We anticipate that this platform will provide new insights into the roles of epigenetic regulation in hematopoiesis, immune cell functions, and immune system aging, and reveal aberrant epigenetic patterns associated with immune-mediated diseases.


Asunto(s)
Cromatina/metabolismo , Epigénesis Genética , Código de Histonas , Histonas/metabolismo , Análisis de la Célula Individual/métodos , Citometría de Flujo , Humanos , Espectrometría de Masas , Procesamiento Proteico-Postraduccional
9.
Nature ; 484(7392): 115-9, 2012 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-22398447

RESUMEN

The recognition of distinctly modified histones by specialized 'effector' proteins constitutes a key mechanism for transducing molecular events at chromatin to biological outcomes. Effector proteins influence DNA-templated processes, including transcription, DNA recombination and DNA repair; however, no effector functions have yet been identified within the mammalian machinery that regulate DNA replication. Here we show that ORC1--a component of ORC (origin of replication complex), which mediates pre-DNA replication licensing--contains a bromo adjacent homology (BAH) domain that specifically recognizes histone H4 dimethylated at lysine 20 (H4K20me2). Recognition of H4K20me2 is a property common to BAH domains present within diverse metazoan ORC1 proteins. Structural studies reveal that the specificity of the BAH domain for H4K20me2 is mediated by a dynamic aromatic dimethyl-lysine-binding cage and multiple intermolecular contacts involving the bound peptide. H4K20me2 is enriched at replication origins, and abrogating ORC1 recognition of H4K20me2 in cells impairs ORC1 occupancy at replication origins, ORC chromatin loading and cell-cycle progression. Mutation of the ORC1 BAH domain has been implicated in the aetiology of Meier-Gorlin syndrome (MGS), a form of primordial dwarfism, and ORC1 depletion in zebrafish results in an MGS-like phenotype. We find that wild-type human ORC1, but not ORC1-H4K20me2-binding mutants, rescues the growth retardation of orc1 morphants. Moreover, zebrafish depleted of H4K20me2 have diminished body size, mirroring the phenotype of orc1 morphants. Together, our results identify the BAH domain as a novel methyl-lysine-binding module, thereby establishing the first direct link between histone methylation and the metazoan DNA replication machinery, and defining a pivotal aetiological role for the canonical H4K20me2 mark, via ORC1, in primordial dwarfism.


Asunto(s)
Replicación del ADN , Trastornos del Crecimiento/metabolismo , Histonas/química , Histonas/metabolismo , Lisina/metabolismo , Micrognatismo/metabolismo , Complejo de Reconocimiento del Origen/química , Complejo de Reconocimiento del Origen/metabolismo , Animales , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Ciclo Celular , Línea Celular , Cromatina/genética , Cromatina/metabolismo , Microtia Congénita , Cristalografía por Rayos X , Replicación del ADN/genética , Modelos Animales de Enfermedad , Enanismo/genética , Enanismo/metabolismo , Oído/anomalías , Trastornos del Crecimiento/genética , Histonas/genética , Humanos , Metilación , Micrognatismo/genética , Modelos Moleculares , Complejo de Reconocimiento del Origen/genética , Rótula/anomalías , Rótula/metabolismo , Estructura Terciaria de Proteína , Origen de Réplica , Pez Cebra , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
10.
Mol Cell ; 33(2): 248-56, 2009 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-19187765

RESUMEN

Aberrations in chromatin dynamics play a fundamental role in tumorigenesis, yet relatively little is known of the molecular mechanisms linking histone lysine methylation to neoplastic disease. ING4 (Inhibitor of Growth 4) is a native subunit of an HBO1 histone acetyltransferase (HAT) complex and a tumor suppressor protein. Here we show a critical role for specific recognition of histone H3 trimethylated at lysine 4 (H3K4me3) by the ING4 PHD finger in mediating ING4 gene expression and tumor suppressor functions. The interaction between ING4 and H3K4me3 augments HBO1 acetylation activity on H3 tails and drives H3 acetylation at ING4 target promoters. Further, ING4 facilitates apoptosis in response to genotoxic stress and inhibits anchorage-independent cell growth, and these functions depend on ING4 interactions with H3K4me3. Together, our results demonstrate a mechanism for brokering crosstalk between H3K4 methylation and H3 acetylation and reveal a molecular link between chromatin modulation and tumor suppressor mechanisms.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Histonas/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Acetilación , Sitios de Unión , Línea Celular Tumoral , Núcleo Celular/metabolismo , Células Cultivadas , Cromatina/metabolismo , Cristalografía por Rayos X , Humanos , Metilación , Modelos Moleculares , Péptidos/química , Péptidos/metabolismo , Conformación Proteica , Especificidad por Sustrato
11.
J Arthroplasty ; 32(9S): S63-S67, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28318869

RESUMEN

A mature national joint registry with widespread adoption and audit can successfully demonstrate trends and influence future orthopedic practice. Correlations can be identified; however, this should not be misinterpreted as causality. It is essential to consider confounding when analyzing observational datasets.


Asunto(s)
Artroplastia , Sistema de Registros , Femenino , Humanos , Masculino
13.
Nature ; 450(7172): 1106-10, 2007 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-18033247

RESUMEN

Nuclear processes such as transcription, DNA replication and recombination are dynamically regulated by chromatin structure. Eukaryotic transcription is known to be regulated by chromatin-associated proteins containing conserved protein domains that specifically recognize distinct covalent post-translational modifications on histones. However, it has been unclear whether similar mechanisms are involved in mammalian DNA recombination. Here we show that RAG2--an essential component of the RAG1/2 V(D)J recombinase, which mediates antigen-receptor gene assembly--contains a plant homeodomain (PHD) finger that specifically recognizes histone H3 trimethylated at lysine 4 (H3K4me3). The high-resolution crystal structure of the mouse RAG2 PHD finger bound to H3K4me3 reveals the molecular basis of H3K4me3-recognition by RAG2. Mutations that abrogate RAG2's recognition of H3K4me3 severely impair V(D)J recombination in vivo. Reducing the level of H3K4me3 similarly leads to a decrease in V(D)J recombination in vivo. Notably, a conserved tryptophan residue (W453) that constitutes a key structural component of the K4me3-binding surface and is essential for RAG2's recognition of H3K4me3 is mutated in patients with immunodeficiency syndromes. Together, our results identify a new function for histone methylation in mammalian DNA recombination. Furthermore, our results provide the first evidence indicating that disrupting the read-out of histone modifications can cause an inherited human disease.


Asunto(s)
Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Reordenamiento Génico de Linfocito B , Histonas/metabolismo , Lisina/metabolismo , Recombinación Genética , VDJ Recombinasas/metabolismo , Secuencias de Aminoácidos , Animales , Sitios de Unión , Histonas/química , Proteínas de Homeodominio/química , Proteínas de Homeodominio/metabolismo , Humanos , Síndromes de Inmunodeficiencia/genética , Lisina/química , Metilación , Ratones , Modelos Moleculares , Unión Proteica , Relación Estructura-Actividad , Especificidad por Sustrato , Triptófano/genética , Triptófano/metabolismo , VDJ Recombinasas/química
14.
J Crohns Colitis ; 17(5): 804-815, 2023 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-36571819

RESUMEN

BACKGROUND AND AIMS: Current understanding of histone post-translational modifications [histone modifications] across immune cell types in patients with inflammatory bowel disease [IBD] during remission and flare is limited. The present study aimed to quantify histone modifications at a single-cell resolution in IBD patients during remission and flare and how they differ compared to healthy controls. METHODS: We performed a case-control study of 94 subjects [83 IBD patients and 11 healthy controls]. IBD patients had either ulcerative colitis [n = 38] or Crohn's disease [n = 45] in clinical remission or flare. We used epigenetic profiling by time-of-flight [EpiTOF] to investigate changes in histone modifications within peripheral blood mononuclear cells from IBD patients. RESULTS: We discovered substantial heterogeneity in histone modifications across multiple immune cell types in IBD patients. They had a higher proportion of less differentiated CD34+ haematopoietic progenitors, and a subset of CD56bright natural killer [NK] cells and γδ T cells characterized by distinct histone modifications associated with gene transcription. The subset of CD56bright NK cells had increases in several histone acetylations. An epigenetically defined subset of NK cells was associated with higher levels of C-reactive protein in peripheral blood. CD34+ monocytes from IBD patients had significantly decreased cleaved H3T22, suggesting they were epigenetically primed for macrophage differentiation. CONCLUSION: We describe the first systems-level quantification of histone modifications across immune cells from IBD patients at a single-cell resolution, revealing the increased epigenetic heterogeneity that is not possible with traditional ChIP-seq profiling. Our data open new directions in investigating the association between histone modifications and IBD pathology using other epigenomic tools.


Asunto(s)
Colitis Ulcerosa , Enfermedades Inflamatorias del Intestino , Humanos , Histonas/metabolismo , Leucocitos Mononucleares/metabolismo , Estudios de Casos y Controles , Procesamiento Proteico-Postraduccional
15.
Cancers (Basel) ; 15(21)2023 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-37958378

RESUMEN

Juvenile myelomonocytic leukemia (JMML) is a deadly pediatric leukemia driven by RAS pathway mutations, of which >35% are gain-of-function in PTPN11. Although DNA hypermethylation portends severe clinical phenotypes, the landscape of histone modifications and chromatin profiles in JMML patient cells have not been explored. Using global mass cytometry, Epigenetic Time of Flight (EpiTOF), we analyzed hematopoietic stem and progenitor cells (HSPCs) from five JMML patients with PTPN11 mutations. These data revealed statistically significant changes in histone methylation, phosphorylation, and acetylation marks that were unique to JMML HSPCs when compared with healthy controls. Consistent with these data, assay for transposase-accessible chromatin with sequencing (ATAC-seq) analysis revealed significant alterations in chromatin profiles at loci encoding post-translational modification enzymes, strongly suggesting their mis-regulated expression. Collectively, this study reveals histone modification pathways as an additional epigenetic abnormality in JMML patient HSPCs, thereby uncovering a new family of potential druggable targets for the treatment of JMML.

16.
JCI Insight ; 8(16)2023 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-37606045

RESUMEN

Systemic lupus erythematosus (SLE) affects 1 in 537 Black women, which is >2-fold more than White women. Black patients develop the disease at a younger age, have more severe symptoms, and have a greater chance of early mortality. We used a multiomics approach to uncover ancestry-associated immune alterations in patients with SLE and healthy controls that may contribute biologically to disease disparities. Cell composition, signaling, epigenetics, and proteomics were evaluated by mass cytometry; droplet-based single-cell transcriptomics and proteomics; and bead-based multiplex soluble mediator levels in plasma. We observed altered whole blood frequencies and enhanced activity in CD8+ T cells, B cells, monocytes, and DCs in Black patients with more active disease. Epigenetic modifications in CD8+ T cells (H3K27ac) could distinguish disease activity level in Black patients and differentiate Black from White patient samples. TLR3/4/7/8/9-related gene expression was elevated in immune cells from Black patients with SLE, and TLR7/8/9 and IFN-α phospho-signaling and cytokine responses were heightened even in immune cells from healthy Black control patients compared with White individuals. TLR stimulation of healthy immune cells recapitulated the ancestry-associated SLE immunophenotypes. This multiomic resource defines ancestry-associated immune phenotypes that differ between Black and White patients with SLE, which may influence the course and severity of SLE and other diseases.


Asunto(s)
Linfocitos B , Lupus Eritematoso Sistémico , Femenino , Humanos , Población Negra , Linfocitos T CD8-positivos , Lupus Eritematoso Sistémico/genética , Fenotipo , Población Blanca
17.
Proc Natl Acad Sci U S A ; 105(41): 15878-83, 2008 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-18840680

RESUMEN

Aire induces ectopic expression of peripheral tissue antigens (PTAs) in thymic medullary epithelial cells, which promotes immunological tolerance. Beginning with a broad screen of histone peptides, we demonstrate that the mechanism by which this single factor controls the transcription of thousands of genes involves recognition of the amino-terminal tail of histone H3, but not of other histones, by one of Aire's plant homeodomain (PHD) fingers. Certain posttranslational modifications of H3 tails, notably dimethylation or trimethylation at H3K4, abrogated binding by Aire, whereas others were tolerated. Similar PHD finger-H3 tail-binding properties were recently reported for BRAF-histone deacetylase complex 80 and DNA methyltransferase 3L; sequence alignment, molecular modeling, and biochemical analyses showed these factors and Aire to have structure-function relationships in common. In addition, certain PHD1 mutations underlying the polyendocrine disorder autoimmune polyendocrinopathy-candidiases-ectodermaldystrophy compromised Aire recognition of H3. In vitro binding assays demonstrated direct physical interaction between Aire and nucleosomes, which was in part buttressed by its affinity to DNA. In vivo Aire interactions with chromosomal regions depleted of H3K4me3 were dependent on its H3 tail-binding activity, and this binding was necessary but not sufficient for the up-regulation of genes encoding PTAs. Thus, Aire's activity as a histone-binding module mediates the thymic display of PTAs that promotes self-tolerance and prevents organ-specific autoimmunity.


Asunto(s)
Histonas/metabolismo , Tolerancia Inmunológica , Dominios y Motivos de Interacción de Proteínas/inmunología , Factores de Transcripción/metabolismo , Animales , Autoantígenos/genética , Autoinmunidad , Cromatina/metabolismo , Ratones , Especificidad de Órganos/inmunología , Unión Proteica , Factores de Transcripción/inmunología , Regulación hacia Arriba , Proteína AIRE
18.
J Med Internet Res ; 13(3): e67, 2011 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-21937354

RESUMEN

Cloud computing is a new way of delivering computing resources and services. Many managers and experts believe that it can improve health care services, benefit health care research, and change the face of health information technology. However, as with any innovation, cloud computing should be rigorously evaluated before its widespread adoption. This paper discusses the concept and its current place in health care, and uses 4 aspects (management, technology, security, and legal) to evaluate the opportunities and challenges of this computing model. Strategic planning that could be used by a health organization to determine its direction, strategy, and resource allocation when it has decided to migrate from traditional to cloud-based health services is also discussed.


Asunto(s)
Redes de Comunicación de Computadores/organización & administración , Computadores , Información de Salud al Consumidor/organización & administración , Almacenamiento y Recuperación de la Información/métodos , Informática en Salud Pública/organización & administración , Integración de Sistemas , Eficiencia Organizacional , Humanos
19.
medRxiv ; 2021 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-33532787

RESUMEN

Coronavirus Disease 2019 (COVID-19), caused by Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2), is associated with a wide range of clinical manifestations, including autoimmune features and autoantibody production. We developed three different protein arrays to measure hallmark IgG autoantibodies associated with Connective Tissue Diseases (CTDs), Anti-Cytokine Antibodies (ACA), and anti-viral antibody responses in 147 hospitalized COVID-19 patients in three different centers. Autoantibodies were identified in approximately 50% of patients, but in <15% of healthy controls. When present, autoantibodies largely targeted autoantigens associated with rare disorders such as myositis, systemic sclerosis and CTD overlap syndromes. Anti-nuclear antibodies (ANA) were observed in ∼25% of patients. Patients with autoantibodies tended to demonstrate one or a few specificities whereas ACA were even more prevalent, and patients often had antibodies to multiple cytokines. Rare patients were identified with IgG antibodies against angiotensin converting enzyme-2 (ACE-2). A subset of autoantibodies and ACA developed de novo following SARS-CoV-2 infection while others were transient. Autoantibodies tracked with longitudinal development of IgG antibodies that recognized SARS-CoV-2 structural proteins such as S1, S2, M, N and a subset of non-structural proteins, but not proteins from influenza, seasonal coronaviruses or other pathogenic viruses. COVID-19 patients with one or more autoantibodies tended to have higher levels of antibodies against SARS-CoV-2 Nonstructural Protein 1 (NSP1) and Methyltransferase (ME). We conclude that SARS-CoV-2 causes development of new-onset IgG autoantibodies in a significant proportion of hospitalized COVID-19 patients and are positively correlated with immune responses to SARS-CoV-2 proteins.

20.
Nat Commun ; 12(1): 5417, 2021 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-34521836

RESUMEN

COVID-19 is associated with a wide range of clinical manifestations, including autoimmune features and autoantibody production. Here we develop three protein arrays to measure IgG autoantibodies associated with connective tissue diseases, anti-cytokine antibodies, and anti-viral antibody responses in serum from 147 hospitalized COVID-19 patients. Autoantibodies are identified in approximately 50% of patients but in less than 15% of healthy controls. When present, autoantibodies largely target autoantigens associated with rare disorders such as myositis, systemic sclerosis and overlap syndromes. A subset of autoantibodies targeting traditional autoantigens or cytokines develop de novo following SARS-CoV-2 infection. Autoantibodies track with longitudinal development of IgG antibodies recognizing SARS-CoV-2 structural proteins and a subset of non-structural proteins, but not proteins from influenza, seasonal coronaviruses or other pathogenic viruses. We conclude that SARS-CoV-2 causes development of new-onset IgG autoantibodies in a significant proportion of hospitalized COVID-19 patients and are positively correlated with immune responses to SARS-CoV-2 proteins.


Asunto(s)
Autoanticuerpos/inmunología , COVID-19/inmunología , Inmunoglobulina G/inmunología , SARS-CoV-2/inmunología , Anciano , Anticuerpos Antinucleares/sangre , Anticuerpos Antinucleares/inmunología , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Autoanticuerpos/sangre , Autoantígenos/inmunología , Enfermedades del Tejido Conjuntivo/inmunología , Citocinas/inmunología , Femenino , Hospitalización , Humanos , Inmunoglobulina G/sangre , Masculino , Persona de Mediana Edad , SARS-CoV-2/patogenicidad , Proteínas Virales/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA