13 research outputs found

    Understanding the relevance of DNA methylation changes in immune differentiation and disease

    Get PDF
    Altres ajuts: We thank CERCA Programme/Generalitat de Catalunya and Josep Carreras Foundation for institutional support. E.B. was funded by [...] and was co-funded by FEDER funds/European Regional Development Fund (ERDF)-a way to build Europe.Immune cells are one of the most complex and diverse systems in the human organism. Such diversity implies an intricate network of different cell types and interactions that are dependently interconnected. The processes by which different cell types differentiate from progenitors, mature, and finally exert their function requires an orchestrated succession of molecular processes that determine cell phenotype and function. The acquisition of these phenotypes is highly dependent on the establishment of unique epigenetic profiles that confer identity and function on the various types of effector cells. These epigenetic mechanisms integrate microenvironmental cues into the genome to establish specific transcriptional programs. Epigenetic modifications bridge environment and genome regulation and play a role in human diseases by their ability to modulate physiological programs through external stimuli. DNA methylation is one of the most ubiquitous, stable, and widely studied epigenetic modifications. Recent technological advances have facilitated the generation of a vast amount of genome-wide DNA methylation data, providing profound insights into the roles of DNA methylation in health and disease. This review considers the relevance of DNA methylation to immune system cellular development and function, as well as the participation of DNA methylation defects in immune-mediated pathologies, illustrated by selected paradigmatic diseases

    Epigenetic and transcriptomic reprogramming in monocytes of severe COVID-19 patients reflects alterations in myeloid differentiation and the influence of inflammatory cytokines

    Get PDF
    COVID-19; Monocytes; Single-cell transcriptomicsCOVID-19; Monocitos; Transcriptómica unicelularCOVID-19; Monòcits; Transcriptòmica unicel·lularBackground COVID-19 manifests with a wide spectrum of clinical phenotypes, ranging from asymptomatic and mild to severe and critical. Severe and critical COVID-19 patients are characterized by marked changes in the myeloid compartment, especially monocytes. However, little is known about the epigenetic alterations that occur in these cells during hyperinflammatory responses in severe COVID-19 patients. Methods In this study, we obtained the DNA methylome and transcriptome of peripheral blood monocytes from severe COVID-19 patients. DNA samples extracted from CD14 + CD15- monocytes of 48 severe COVID-19 patients and 11 healthy controls were hybridized on MethylationEPIC BeadChip arrays. In parallel, single-cell transcriptomics of 10 severe COVID-19 patients were generated. CellPhoneDB was used to infer changes in the crosstalk between monocytes and other immune cell types. Results We observed DNA methylation changes in CpG sites associated with interferon-related genes and genes associated with antigen presentation, concordant with gene expression changes. These changes significantly overlapped with those occurring in bacterial sepsis, although specific DNA methylation alterations in genes specific to viral infection were also identified. We also found these alterations to comprise some of the DNA methylation changes occurring during myeloid differentiation and under the influence of inflammatory cytokines. A progression of DNA methylation alterations in relation to the Sequential Organ Failure Assessment (SOFA) score was found to be related to interferon-related genes and T-helper 1 cell cytokine production. CellPhoneDB analysis of the single-cell transcriptomes of other immune cell types suggested the existence of altered crosstalk between monocytes and other cell types like NK cells and regulatory T cells. Conclusion Our findings show the occurrence of an epigenetic and transcriptional reprogramming of peripheral blood monocytes, which could be associated with the release of aberrant immature monocytes, increased systemic levels of pro-inflammatory cytokines, and changes in immune cell crosstalk in these patients.This study has been funded by R+D+i project of the Spanish Ministry of Science and Innovation (grant number PID2020-117212RB-I00/ MICIN/AEI/10.13039/501100011033). We also thank the Chan Zuckerberg Initiative (grant 2020–216799) and Wellcome Sanger core funding (WT206194). This publication has also been supported by the Unstoppable campaign of the Josep Carreras Leukaemia Foundation. Additional funding comes from Project PI18/00346 (Instituto de Salud Carlos III and co-funded by European Union (ERDF/ESF ). A.B. received additional support from a Gates Cambridge Scholarship

    Targeting aberrant DNA methylation in mesenchymal stromal cells as a treatment for myeloma bone disease

    Get PDF
    Multiple myeloma (MM) progression and myeloma-associated bone disease (MBD) are highly dependent on bone marrow mesenchymal stromal cells (MSCs). MM-MSCs exhibit abnormal transcriptomes, suggesting the involvement of epigenetic mechanisms governing their tumor-promoting functions and prolonged osteoblast suppression. Here, we identify widespread DNA methylation alterations of bone marrow-isolated MSCs from distinct MM stages, particularly in Homeobox genes involved in osteogenic differentiation that associate with their aberrant expression. Moreover, these DNA methylation changes are recapitulated in vitro by exposing MSCs from healthy individuals to MM cells. Pharmacological targeting of DNMTs and G9a with dual inhibitor CM-272 reverts the expression of hypermethylated osteogenic regulators and promotes osteoblast differentiation of myeloma MSCs. Most importantly, CM-272 treatment prevents tumor-associated bone loss and reduces tumor burden in a murine myeloma model. Our results demonstrate that epigenetic aberrancies mediate the impairment of bone formation in MM, and its targeting by CM-272 is able to reverse MBD. Mesenchymal stromal cells (MSCs) have been shown to support multiple myeloma (MM) development. Here, MSCs isolated from the bone marrow of MM patients are shown to have altered DNA methylation patterns and a methyltransferase inhibitor reverts MM-associated bone loss and reduces tumour burden in MM murine models

    The synovial and blood monocyte DNA methylomes mirror prognosis, evolution, and treatment in early arthritis

    Get PDF
    Identifying predictive biomarkers at early stages of inflammatory arthritis is crucial for starting appropriate therapies to avoid poor outcomes. Monocytes (MOs) and macrophages, largely associated with arthritis, are contributors and sensors of inflammation through epigenetic modifications. In this study, we investigated associations between clinical features and DNA methylation in blood and synovial fluid (SF) MOs in a prospective cohort of patients with early inflammatory arthritis. DNA methylation profiles of undifferentiated arthritis (UA) blood MOs exhibited marked alterations in comparison with those from healthy donors. We identified additional differences both in blood and SF MOs after comparing patients with UA grouped by their future outcomes, i.e., good versus poor. Patient profiles in subsequent visits revealed a reversion toward a healthy level in both groups, those requiring disease-modifying antirheumatic drugs and those who remitted spontaneously. Changes in disease activity between visits also affected DNA methylation, which was partially concomitant in the SF of UA and in blood MOs of patients with rheumatoid arthritis. Epigenetic similarities between arthritis types allow a common prediction of disease activity. Our results constitute a resource of DNA methylation-based biomarkers of poor prognosis, disease activity, and treatment efficacy for the personalized clinical management of early inflammatory arthritis.We thank CERCA Programme/Generalitat de Catalunya and the Josep Carreras Foundation for institutional support. The authors thank all the patients who graciously donated their time and samples to further arthritis research. We are also thankful to Núria Sapena, Marta Bassas, and Cristina González, nurses from the outpatient clinic of the Department of Rheumatology, for their help in the management of biologic samples. This research was funded by Fondo de Investigación en Salud (FIS) grant PI17/00993 from the Institute of Health Carlos III (ISCIII) (to JDC); by grants SAF2017-88086-R and PID2020-117212RB-I00 / AEI / 10.13038/501100011033) from the Spanish Ministry of Science and Innovation (MICINN) (to EB); and by the Thematic Networks for Cooperative Research (RETICS) grant provided by ISCII, Research Network for Inflammation and Rheumatic Diseases (RIER) RD16/0012/0013, cofinanced by the European Fund for Regional Development’s (FEDER) Una manera de hacer Europa program (to JDC and EB).Peer reviewe

    COVID-19 progression and convalescence in common variable immunodeficiency patients shows incomplete adaptive responses and persistent inflammasome activation

    Get PDF
    Patients with common variable immunodeficiency (CVID), the most prevalent symptomatic primary immunodeficiency, are characterized by hypogammaglobulinemia, poorly protective vaccine titers and increased susceptibility to infections. New pathogens such as the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease 2019 (COVID-19), might constitute a particular threat to these immunocompromised patients since many of them experience a slower recovery and do not achieve full response to SARS-CoV-2 vaccines. To define the molecular basis of the altered immune responses caused by SARS-CoV-2 infection in CVID patients, we generated longitudinal single-cell datasets of peripheral blood immune cells along viral infection and recovery. We sampled the same individuals before, during and after SARS-CoV-2 infection to model their specific immune response dynamics while removing donor variability. We observed that COVID-19 CVID patients show defective canonical NF-κB pathway activation and dysregulated expression of BCR-related genes in naïve B cells, as well as enhanced cytotoxic activity but incomplete cytokine response in NK and T cells. Moreover, monocytes from COVID-19 CVID patients show persistent activation of several inflammasome-related genes, including the pyrin and NLRC4 inflammasomes. Our results shed light on the molecular basis of the prolonged clinical manifestations observed in these immunodeficient patients upon SARS-CoV-2 infection, which might illuminate the development of tailored treatments for COVID-19 CVID patients.We thank the CERCA Program/Generalitat de Catalunya and the Josep Carreras Foundation for institutional support. This publication is part of the Human Cell Atlas: www.humancellatlas.org/publications. This study was funded by ”la Caixa” Foundation under the grant agreement LCF/PR/HR22/52420002, Spanish Ministry of Science and Innovation (grant number PID2020-117212RB-I00/AEI/10.13038/501100011033) (E.B.), by the Wellcome Trust Grant 206194 and 108413/A/15/D (R.V.-T.), Instituto de Salud Carlos III (ISCIII), Ref. AC18/00057, associated with i-PAD project (ERARE European Union program) (E.B.), and the Chan Zuckerberg Initiative (grant 2020-216799) (R.V.-T. and E.B.). This publication has also been supported by the Unstoppable campaign of the Josep Carreras Leukaemia Foundation. We are indebted to the donors for participating in this research.N

    Single-cell Atlas of common variable immunodeficiency shows germinal center-associated epigenetic dysregulation in B-cell responses

    Get PDF
    Common variable immunodeficiency (CVID), the most prevalent symptomatic primary immunodeficiency, displays impaired terminal B-cell differentiation and defective antibody responses. Incomplete genetic penetrance and ample phenotypic expressivity in CVID suggest the participation of additional pathogenic mechanisms. Monozygotic (MZ) twins discordant for CVID are uniquely valuable for studying the contribution of epigenetics to the disease. Here, we generate a single-cell epigenomics and transcriptomics census of naïve-to-memory B cell differentiation in a CVID-discordant MZ twin pair. Our analysis identifies DNA methylation, chromatin accessibility and transcriptional defects in memory B-cells mirroring defective cell-cell communication upon activation. These findings are validated in a cohort of CVID patients and healthy donors. Our findings provide a comprehensive multi-omics map of alterations in naïve-to-memory B-cell transition in CVID and indicate links between the epigenome and immune cell cross-talk. Our resource, publicly available at the Human Cell Atlas, gives insight into future diagnosis and treatments of CVID patients

    Epigenetic Dysregulation of Monocytes as a Sensor of Activity and Progression in Arthritis

    Full text link
    [eng] OBJECTIVE: Identifying predictive biomarkers at early stages of undifferentiated arthritis (UA) is crucial for starting appropriate therapies to avoid poor outcomes. Monocytes and macrophages, largely associated with arthritis, are contributors and sensors of inflammation through epigenetic modifications. We investigated associations between clinical features and DNA methylation patterns in monocytes from UA patients, seeking potential prognostic biomarkers. METHODS: We characterized the DNA methylation profiles of blood and synovial fluid (SF) monocytes from UA patients at baseline and during subsequent visits and compared them to those from healthy donors (HD) and in vitro-differentiated macrophages. These data were used to interrogate their relationship with prognosis and changes over time, and identify predictive biomarkers of poor prognosis. RESULTS: UA blood monocyte profiles exhibited significant alterations in comparison with those from HD. After comparing UA patients grouped by their future outcomes —good versus poor—, additional differences were identified both in blood and SF monocytes. Patient profiles in subsequent visits revealed a reversion towards a healthy level in both groups, those requiring DMARD treatment and those that remitted spontaneously. Changes in disease activity between visits revealed an impact on DNA methylation, partially concomitant in the SF of UA and in blood monocytes of rheumatoid arthritis patients. CONCLUSION: We propose a list of DNA methylation-based biomarkers of poor prognosis in early untreated UA patients. We demonstrate the value of DNA methylation as a tool for estimating disease activity in UA. Finally, we highlight epigenetic similarities between arthritis types that allow a common prediction of disease activity.[spa] OBJETIVO: La identificación de biomarcadores predictivos en fases tempranas de la artritis indiferenciada (AI) es crucial para iniciar las terapias apropiadas para evitar malos resultados. Los monocitos y macrófagos, asociados en gran medida a la artritis, son contribuyentes y sensores de la inflamación a través de modificaciones epigenéticas. Investigamos las asociaciones entre las características clínicas y los patrones de metilación del ADN en monocitos de pacientes con AI, buscando potenciales biomarcadores de pronóstico. MÉTODOS: Caracterizamos los perfiles de metilación del ADN de los monocitos de sangre y líquido sinovial (LS) de pacientes con AI al inicio y durante las visitas posteriores y los comparamos con los de donantes sanos (DS) y macrófagos diferenciados in vitro. Estos datos se utilizaron para interrogar su relación con el pronóstico y los cambios a lo largo del tiempo, e identificar biomarcadores predictivos de mal pronóstico. RESULTADOS: Los perfiles de monocitos en sangre de la AI mostraron alteraciones significativas en comparación con los de la EH. Tras comparar a los pacientes de AU agrupados por sus resultados futuros -buenos frente a malos-, se identificaron diferencias adicionales tanto en los monocitos sanguíneos como en los de SF. Los perfiles de los pacientes en las visitas posteriores revelaron una reversión hacia un nivel saludable en ambos grupos, los que requirieron tratamiento con DMARD y los que remitieron espontáneamente. Los cambios en la actividad de la enfermedad entre las visitas revelaron un impacto en la metilación del ADN, parcialmente concomitante en el SF de la UA y en los monocitos sanguíneos de los pacientes con artritis reumatoide. CONCLUSIONES: Proponemos una lista de biomarcadores basados en la metilación del ADN de mal pronóstico en pacientes con AI no tratados en fase inicial. Demostramos el valor de la metilación del ADN como herramienta para estimar la actividad de la enfermedad en la AI. Por último, destacamos las similitudes epigenéticas entre los tipos de artritis que permiten una predicción común de la actividad de la enfermedad

    Understanding the relevance of DNA methylation changes in immune differentiation and disease

    Get PDF
    Altres ajuts: We thank CERCA Programme/Generalitat de Catalunya and Josep Carreras Foundation for institutional support. E.B. was funded by [...] and was co-funded by FEDER funds/European Regional Development Fund (ERDF)-a way to build Europe.Immune cells are one of the most complex and diverse systems in the human organism. Such diversity implies an intricate network of different cell types and interactions that are dependently interconnected. The processes by which different cell types differentiate from progenitors, mature, and finally exert their function requires an orchestrated succession of molecular processes that determine cell phenotype and function. The acquisition of these phenotypes is highly dependent on the establishment of unique epigenetic profiles that confer identity and function on the various types of effector cells. These epigenetic mechanisms integrate microenvironmental cues into the genome to establish specific transcriptional programs. Epigenetic modifications bridge environment and genome regulation and play a role in human diseases by their ability to modulate physiological programs through external stimuli. DNA methylation is one of the most ubiquitous, stable, and widely studied epigenetic modifications. Recent technological advances have facilitated the generation of a vast amount of genome-wide DNA methylation data, providing profound insights into the roles of DNA methylation in health and disease. This review considers the relevance of DNA methylation to immune system cellular development and function, as well as the participation of DNA methylation defects in immune-mediated pathologies, illustrated by selected paradigmatic diseases

    Coordinated glucocorticoid receptor and MAFB action induces tolerogenesis and epigenome remodeling in dendritic cells

    Get PDF
    Glucocorticoids (GCs) exert potent anti-inflammatory effects in immune cells through the glucocorticoid receptor (GR). Dendritic cells (DCs), central actors for coordinating immune responses, acquire tolerogenic properties in response to GCs. Tolerogenic DCs (tolDCs) have emerged as a potential treatment for various inflammatory diseases. To date, the underlying cell type-specific regulatory mechanisms orchestrating GC-mediated acquisition of immunosuppressive properties remain poorly understood. In this study, we investigated the transcriptomic and epigenomic remodeling associated with differentiation to DCs in the presence of GCs. Our analysis demonstrates a major role of MAFB in this process, in synergy with GR. GR and MAFB both interact with methylcytosine dioxygenase TET2 and bind to genomic loci that undergo specific demethylation in tolDCs. We also show that the role of MAFB is more extensive, binding to thousands of genomic loci in tolDCs. Finally, MAFB knockdown erases the tolerogenic properties of tolDCs and reverts the specific DNA demethylation and gene upregulation. The preeminent role of MAFB is also demonstrated in vivo for myeloid cells from synovium in rheumatoid arthritis following GC treatment. Our results imply that, once directly activated by GR, MAFB plays a critical role in orchestrating the epigenomic and transcriptomic remodeling that define the tolerogenic phenotype

    Epigenetic and transcriptomic reprogramming in monocytes of severe COVID-19 patients reflects alterations in myeloid differentiation and the influence of inflammatory cytokines.

    Get PDF
    BACKGROUND: COVID-19 manifests with a wide spectrum of clinical phenotypes, ranging from asymptomatic and mild to severe and critical. Severe and critical COVID-19 patients are characterized by marked changes in the myeloid compartment, especially monocytes. However, little is known about the epigenetic alterations that occur in these cells during hyperinflammatory responses in severe COVID-19 patients. METHODS: In this study, we obtained the DNA methylome and transcriptome of peripheral blood monocytes from severe COVID-19 patients. DNA samples extracted from CD14 + CD15- monocytes of 48 severe COVID-19 patients and 11 healthy controls were hybridized on MethylationEPIC BeadChip arrays. In parallel, single-cell transcriptomics of 10 severe COVID-19 patients were generated. CellPhoneDB was used to infer changes in the crosstalk between monocytes and other immune cell types. RESULTS: We observed DNA methylation changes in CpG sites associated with interferon-related genes and genes associated with antigen presentation, concordant with gene expression changes. These changes significantly overlapped with those occurring in bacterial sepsis, although specific DNA methylation alterations in genes specific to viral infection were also identified. We also found these alterations to comprise some of the DNA methylation changes occurring during myeloid differentiation and under the influence of inflammatory cytokines. A progression of DNA methylation alterations in relation to the Sequential Organ Failure Assessment (SOFA) score was found to be related to interferon-related genes and T-helper 1 cell cytokine production. CellPhoneDB analysis of the single-cell transcriptomes of other immune cell types suggested the existence of altered crosstalk between monocytes and other cell types like NK cells and regulatory T cells. CONCLUSION: Our findings show the occurrence of an epigenetic and transcriptional reprogramming of peripheral blood monocytes, which could be associated with the release of aberrant immature monocytes, increased systemic levels of pro-inflammatory cytokines, and changes in immune cell crosstalk in these patients
    corecore