Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Eur J Immunol ; 54(1): e2350633, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37799110

ABSTRACT

In COVID-19, hyperinflammatory and dysregulated immune responses contribute to severity. Patients with pre-existing autoimmune conditions can therefore be at increased risk of severe COVID-19 and/or associated sequelae, yet SARS-CoV-2 infection in this group has been little studied. Here, we performed single-cell analysis of peripheral blood mononuclear cells from patients with three major autoimmune diseases (rheumatoid arthritis, psoriasis, or multiple sclerosis) during SARS-CoV-2 infection. We observed compositional differences between the autoimmune disease groups coupled with altered patterns of gene expression, transcription factor activity, and cell-cell communication that substantially shape the immune response under SARS-CoV-2 infection. While enrichment of HLA-DRlow CD14+ monocytes was observed in all three autoimmune disease groups, type-I interferon signaling as well as inflammatory T cell and monocyte responses varied widely between the three groups of patients. Our results reveal disturbed immune responses to SARS-CoV-2 in patients with pre-existing autoimmunity, highlighting important considerations for disease treatment and follow-up.


Subject(s)
Autoimmune Diseases , COVID-19 , Humans , SARS-CoV-2 , Leukocytes, Mononuclear , Multiomics , Autoimmunity , Single-Cell Analysis
2.
Rev. esp. quimioter ; 36(2): 187-193, abr. 2023. ilus, tab, graf
Article in English | IBECS | ID: ibc-217400

ABSTRACT

Introduction: This study aimed to evaluate whether early vitamin C and thiamine administration was associated with a lower 28-day and in-hospital mortality in surgical critically ill patients with refractory septic shock. Patients and methods: We performed a retrospective before-and-after study on patients with refractory septic shock. According to local protocol, hydrocortisone is initiated in case of refractory septic shock. In January 2017, the protocol was changed and vitamin C and thiamine were included. Patients who were admitted in 2015-2016 and 2017-2018 were included in the control and treatment groups, respectively. The primary end point was 28-day and in-hospital mortality. Secondary end points were ICU mortality, ICU and hospital length of stay, duration of vasopressors and mechanical ventilation, use of renal replacement therapy (RRT), and the modification in serum procalcitonin and SOFA score during the first 72 h. Results: A total of 120 patients were included (58 in the treatment group and 62 in the control group). Log-rank test in Kaplan-Meier curves showed lower 28-day and in-hospital mortality over time in the treatment group (p=0.021 and p=0.035, respectively) but it not reached statistical significance in ICU mortality over time (p=0.100). The need of RRT was less frequent in treatment group (17.2% vs. 37.1%, p=0.024). There were no differences in other secondary outcomes. Conclusions: Intravenous vitamin C and thiamine administration in surgical patients with refractory septic shock may be associated with a lower 28-day and in-hospital mortality. Further prospective studies are needed in refractory septic shock. (AU)


Introducción: El objetivo de este estudio fue evaluar si la administración precoz de vitamina C y tiamina estaba asociada a una reducción en la mortalidad a los 28 días y hospitalaria en pacientes críticos quirúrgicos con shock séptico refractario. Pacientes y métodos: Realizamos un estudio retrospectivo antes-después en pacientes con shock séptico refractario. Según el protocolo local, se inicia tratamiento con hidrocortisona en situación de shock séptico refractario. En enero de 2017 se cambió el protocolo y se incluyó vitamina C y tiamina. Los pacientes que fueron ingresados en 2015-2016 y 2017-2018 se incluyeron en el grupo control y tratamiento, respectivamente. Los objetivos primarios fueron la mortalidad a los 28 días y hospitalaria. Los objetivos secundarios fueron la mortalidad en UCI, la duración de estancia en UCI y hospitalaria, la duración del tratamiento vasopresor y de la ventilación mecánica, el uso de técnicas de reemplazo renal (TRR), y la modificación en la procalcitonina sérica y la puntuación SOFA durante las primeras 72h. Resultados: Se incluyeron un total de 120 pacientes (58 en el grupo tratamiento y 62 en el grupo control). El test Log-rank en las curvas de Kaplan-Meier mostró mortalidad a los 28 días y hospitalaria más baja a lo largo del tiempo en el grupo tratamiento (p=0,021 and p=0,035, respectivamente) pero no alcanzó significación estadística en la mortalidad en UCI a lo largo del tiempo (p=0,100). La necesidad de TRR fue menos frecuente en el grupo tratamiento (17,2% vs. 37,1%, p=0,024). No hubo diferencias en otros resultados secundarios. Conclusiones: La administración de vitamina C y tiamina intravenosa en pacientes quirúrgicos con shock séptico refractario podría estar asociada a una menor mortalidad a los 28 días y hospitalaria. Se necesitan más estudios prospectivos en pacientes con shock séptico refractario. (AU)


Subject(s)
Humans , Male , Female , Middle Aged , Aged , Aged, 80 and over , Vitamin D/therapeutic use , Thiamine/therapeutic use , Shock, Septic/drug therapy , Retrospective Studies , Vasoconstrictor Agents
3.
Clin Epigenetics ; 14(1): 188, 2022 12 28.
Article in English | MEDLINE | ID: mdl-36575526

ABSTRACT

BACKGROUND: DNA methylation profiling of circulating cell-free DNA (cfDNA) has rapidly become a promising strategy for biomarker identification and development. The cell-type-specific nature of DNA methylation patterns and the direct relationship between cfDNA and apoptosis can potentially be used non-invasively to predict local alterations. In addition, direct detection of altered DNA methylation patterns performs well as a biomarker. In a previous study, we demonstrated marked DNA methylation alterations in brain tissue from patients with mesial temporal lobe epilepsy with hippocampal sclerosis (MTLE-HS). RESULTS: We performed DNA methylation profiling in cfDNA isolated from the serum of MTLE patients and healthy controls using BeadChip arrays followed by systematic bioinformatic analysis including deconvolution analysis and integration with DNase accessibility data sets. Differential cfDNA methylation analysis showed an overrepresentation of gene ontology terms and transcription factors related to central nervous system function and regulation. Deconvolution analysis of the DNA methylation data sets ruled out the possibility that the observed differences were due to changes in the proportional contribution of cortical neurons in cfDNA. Moreover, we found no overrepresentation of neuron- or glia-specific patterns in the described cfDNA methylation patterns. However, the MTLE-HS cfDNA methylation patterns featured a significant overrepresentation of the epileptic DNA methylation alterations previously observed in the hippocampus. CONCLUSIONS: Our results support the use of cfDNA methylation profiling as a rational approach to seeking non-invasive and reproducible epilepsy biomarkers.


Subject(s)
Cell-Free Nucleic Acids , Epilepsy, Temporal Lobe , Humans , DNA Methylation , Hippocampus , Epilepsy, Temporal Lobe/genetics , Brain , Cell-Free Nucleic Acids/genetics
4.
Genome Med ; 14(1): 134, 2022 11 29.
Article in English | MEDLINE | ID: mdl-36443794

ABSTRACT

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.


Subject(s)
COVID-19 , Monocytes , Humans , Transcriptome , Cytokines , COVID-19/genetics , Interferons , Antiviral Agents , Epigenesis, Genetic
5.
Nucleic Acids Res ; 50(19): 10981-10994, 2022 10 28.
Article in English | MEDLINE | ID: mdl-36305821

ABSTRACT

Dendritic cells (DCs), the most potent antigen-presenting cells, are necessary for effective activation of naïve T cells. DCs' immunological properties are modulated in response to various stimuli. Active DNA demethylation is crucial for DC differentiation and function. Vitamin C, a known cofactor of ten-eleven translocation (TET) enzymes, drives active demethylation. Vitamin C has recently emerged as a promising adjuvant for several types of cancer; however, its effects on human immune cells are poorly understood. In this study, we investigate the epigenomic and transcriptomic reprogramming orchestrated by vitamin C in monocyte-derived DC differentiation and maturation. Vitamin C triggers extensive demethylation at NF-κB/p65 binding sites, together with concordant upregulation of antigen-presentation and immune response-related genes during DC maturation. p65 interacts with TET2 and mediates the aforementioned vitamin C-mediated changes, as demonstrated by pharmacological inhibition. Moreover, vitamin C increases TNFß production in DCs through NF-κB, in concordance with the upregulation of its coding gene and the demethylation of adjacent CpGs. Finally, vitamin C enhances DC's ability to stimulate the proliferation of autologous antigen-specific T cells. We propose that vitamin C could potentially improve monocyte-derived DC-based cell therapies.


Subject(s)
Ascorbic Acid , Dendritic Cells , Epigenesis, Genetic , NF-kappa B , Humans , Ascorbic Acid/pharmacology , Cell Differentiation/genetics , NF-kappa B/metabolism , T-Lymphocytes/metabolism , Cellular Reprogramming
6.
Ann Rheum Dis ; 2022 Jun 15.
Article in English | MEDLINE | ID: mdl-35705375

ABSTRACT

OBJECTIVES: Giant cell arteritis (GCA) is a complex systemic vasculitis mediated by the interplay between both genetic and epigenetic factors. Monocytes are crucial players of the inflammation occurring in GCA. Therefore, characterisation of the monocyte methylome and transcriptome in GCA would be helpful to better understand disease pathogenesis. METHODS: We performed an integrated epigenome-and transcriptome-wide association study in CD14+ monocytes from 82 patients with GCA, cross-sectionally classified into three different clinical statuses (active, in remission with or without glucocorticoid (GC) treatment), and 31 healthy controls. RESULTS: We identified a global methylation and gene expression dysregulation in GCA monocytes. Specifically, monocytes from active patients showed a more proinflammatory phenotype compared with healthy controls and patients in remission. In addition to inflammatory pathways known to be involved in active GCA, such as response to IL-6 and IL-1, we identified response to IL-11 as a new pathway potentially implicated in GCA. Furthermore, monocytes from patients in remission with treatment showed downregulation of genes involved in inflammatory processes as well as overexpression of GC receptor-target genes. Finally, we identified changes in DNA methylation correlating with alterations in expression levels of genes with a potential role in GCA pathogenesis, such as ITGA7 and CD63, as well as genes mediating the molecular response to GC, including FKBP5, ETS2, ZBTB16 and ADAMTS2. CONCLUSION: Our results revealed profound alterations in the methylation and transcriptomic profiles of monocytes from GCA patients, uncovering novel genes and pathways involved in GCA pathogenesis and in the molecular response to GC treatment.

7.
Nat Commun ; 13(1): 1779, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35365635

ABSTRACT

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.


Subject(s)
Common Variable Immunodeficiency , B-Lymphocytes , Common Variable Immunodeficiency/diagnosis , Common Variable Immunodeficiency/genetics , Epigenesis, Genetic , Epigenomics , Germinal Center , Humans
8.
JCI Insight ; 7(9)2022 05 09.
Article in English | MEDLINE | ID: mdl-35324478

ABSTRACT

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.


Subject(s)
Arthritis, Rheumatoid , Epigenome , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/genetics , Biomarkers , Humans , Monocytes , Prognosis , Prospective Studies , Synovial Membrane
9.
Cell Rep ; 38(3): 110244, 2022 01 18.
Article in English | MEDLINE | ID: mdl-35045292

ABSTRACT

The active form of vitamin D, 1,25-dihydroxyvitamin D3, induces a stable tolerogenic phenotype in dendritic cells (DCs). This process involves the vitamin D receptor (VDR), which translocates to the nucleus, binds its cognate genomic sites, and promotes epigenetic and transcriptional remodeling. In this study, we report the occurrence of vitamin D-specific DNA demethylation and transcriptional activation at VDR binding sites associated with the acquisition of tolerogenesis in vitro. Differentiation to tolerogenic DCs associates with activation of the IL-6-JAK-STAT3 pathway. We show that JAK2-mediated STAT3 phosphorylation is specific to vitamin D stimulation. VDR and the phosphorylated form of STAT3 interact with each other to form a complex with methylcytosine dioxygenase TET2. Most importantly, pharmacological inhibition of JAK2 reverts vitamin D-induced tolerogenic properties of DCs. This interplay among VDR, STAT3, and TET2 opens up possibilities for modulating DC immunogenic properties in clinics.


Subject(s)
DNA-Binding Proteins/immunology , Dendritic Cells/immunology , Dioxygenases/immunology , Immune Tolerance/immunology , Receptors, Calcitriol/immunology , STAT3 Transcription Factor/immunology , Cells, Cultured , DNA-Binding Proteins/metabolism , Dendritic Cells/metabolism , Dioxygenases/metabolism , Humans , Receptors, Calcitriol/metabolism , STAT3 Transcription Factor/metabolism
10.
Prog Neurobiol ; 209: 102207, 2022 02.
Article in English | MEDLINE | ID: mdl-34923048

ABSTRACT

Mesial temporal lobe epilepsy with hippocampal sclerosis (MTLE-HS) is the most common focal epilepsy in adults. It is characterized by alarming rates of pharmacoresistance. Epileptogenesis is associated with the occurrence of epigenetic alterations, and the few epigenetic studies carried out in MTLE-HS have mainly focused on the hippocampus. In this study, we obtained the DNA methylation profiles from both the hippocampus and anterior temporal neocortex of MTLE-HS patients subjected to resective epilepsy surgery and autopsied non-epileptic controls. We assessed the progressive nature of DNA methylation changes in relation to epilepsy duration. We identified significantly altered hippocampal DNA methylation patterns encompassing multiple pathways known to be involved in epileptogenesis. DNA methylation changes were even more striking in the neocortex, wherein pathogenic pathways and genes were common to both tissues. Most importantly, DNA methylation changes at many genomic sites varied significantly with epilepsy duration. Such progressive changes were associated with inflammation-related genes in the hippocampus. Our results suggest that the neocortex, relatively spared of extensive histopathological damage, may also be involved in epilepsy development. These results also open the possibility that the observed neocortical impairment could represent a preliminary stage of epileptogenesis before the establishment of chronic lesions or a consequence of prolonged seizure exposure. Our two-tissue multi-level characterization of the MTLE-HS DNA methylome suggests the occurrence of a self-propagating inflammatory wave of epigenetic dysregulation.


Subject(s)
Epilepsy, Temporal Lobe , Epilepsy , Adult , DNA Methylation/genetics , Epilepsy, Temporal Lobe/genetics , Hippocampus/pathology , Humans , Sclerosis/complications , Sclerosis/pathology
11.
Nucleic Acids Res ; 50(1): 108-126, 2022 01 11.
Article in English | MEDLINE | ID: mdl-34893889

ABSTRACT

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.


Subject(s)
Dendritic Cells/immunology , Epigenesis, Genetic , Immune Tolerance , MafB Transcription Factor/metabolism , Receptors, Glucocorticoid/metabolism , Adult , Cells, Cultured , DNA Methylation , DNA-Binding Proteins/metabolism , Dioxygenases/metabolism , Female , Humans , MafB Transcription Factor/genetics , Male , Middle Aged
12.
Front Immunol ; 12: 734652, 2021.
Article in English | MEDLINE | ID: mdl-34867954

ABSTRACT

Microbial challenges, such as widespread bacterial infection in sepsis, induce endotoxin tolerance, a state of hyporesponsiveness to subsequent infections. The participation of DNA methylation in this process is poorly known. In this study, we perform integrated analysis of DNA methylation and transcriptional changes following in vitro exposure to gram-negative bacterial lipopolysaccharide, together with analysis of ex vivo monocytes from septic patients. We identify TET2-mediated demethylation and transcriptional activation of inflammation-related genes that is specific to toll-like receptor stimulation. Changes also involve phosphorylation of STAT1, STAT3 and STAT5, elements of the JAK2 pathway. JAK2 pathway inhibition impairs the activation of tolerized genes on the first encounter with lipopolysaccharide. We then confirm the implication of the JAK2-STAT pathway in the aberrant DNA methylome of patients with sepsis caused by gram-negative bacteria. Finally, JAK2 inhibition in monocytes partially recapitulates the expression changes produced in the immunosuppressive cellular state acquired by monocytes from gram-negative sepsis, as described by single cell-RNA-sequencing. Our study evidences both the crucial role the JAK2-STAT pathway in epigenetic regulation and initial response of the tolerized genes to gram-negative bacterial endotoxins and provides a pharmacological target to prevent exacerbated responses.


Subject(s)
Endotoxin Tolerance/genetics , Gram-Negative Bacteria/immunology , Gram-Negative Bacterial Infections/genetics , Gram-Negative Bacterial Infections/immunology , Monocytes/immunology , Monocytes/microbiology , Sepsis/genetics , Sepsis/immunology , Case-Control Studies , DNA Methylation/genetics , DNA Methylation/immunology , Endotoxin Tolerance/drug effects , Endotoxin Tolerance/immunology , Endotoxins/toxicity , Epigenesis, Genetic , Female , Gram-Negative Bacterial Infections/microbiology , Humans , In Vitro Techniques , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/genetics , Janus Kinase 2/immunology , Lipopolysaccharides/toxicity , Male , Monocytes/drug effects , STAT Transcription Factors/genetics , STAT Transcription Factors/immunology , Sepsis/microbiology , Signal Transduction/genetics , Signal Transduction/immunology , Toll-Like Receptor 2/immunology , Toll-Like Receptor 4/immunology
13.
Nucleic Acids Res ; 49(9): 5057-5073, 2021 05 21.
Article in English | MEDLINE | ID: mdl-33950194

ABSTRACT

Activation-induced deaminase (AID) initiates antibody diversification in germinal center B cells by deaminating cytosines, leading to somatic hypermutation and class-switch recombination. Loss-of-function mutations in AID lead to hyper-IgM syndrome type 2 (HIGM2), a rare human primary antibody deficiency. AID-mediated deamination has been proposed as leading to active demethylation of 5-methycytosines in the DNA, although evidence both supports and casts doubt on such a role. In this study, using whole-genome bisulfite sequencing of HIGM2 B cells, we investigated direct AID involvement in active DNA demethylation. HIGM2 naïve and memory B cells both display widespread DNA methylation alterations, of which ∼25% are attributable to active DNA demethylation. For genes that undergo active demethylation that is impaired in HIGM2 individuals, our analysis indicates that AID is not directly involved. We demonstrate that the widespread alterations in the DNA methylation and expression profiles of HIGM2 naïve B cells result from premature overstimulation of the B-cell receptor prior to the germinal center reaction. Our data support a role for AID in B cell central tolerance in preventing the expansion of autoreactive cell clones, affecting the correct establishment of DNA methylation patterns.


Subject(s)
B-Lymphocytes/immunology , Cytidine Deaminase/physiology , DNA Methylation , Hyper-IgM Immunodeficiency Syndrome/genetics , Hyper-IgM Immunodeficiency Syndrome/immunology , Autoimmunity , B-Lymphocytes/metabolism , Cytidine Deaminase/deficiency , Cytidine Deaminase/genetics , Germinal Center/immunology , Humans , Hyper-IgM Immunodeficiency Syndrome/metabolism , Immune Tolerance , Immunologic Memory , Receptors, Antigen, B-Cell/genetics , Transcriptome , Whole Genome Sequencing
14.
Nat Commun ; 12(1): 421, 2021 01 18.
Article in English | MEDLINE | ID: mdl-33462210

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/pharmacology , Bone Diseases/drug therapy , DNA Methylation/drug effects , Enzyme Inhibitors/pharmacology , Mesenchymal Stem Cells/drug effects , Multiple Myeloma/drug therapy , Adult , Aged , Aged, 80 and over , Animals , Antineoplastic Agents/therapeutic use , Bone Diseases/diagnosis , Bone Diseases/genetics , Bone Diseases/pathology , Bone Marrow/pathology , DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , DNA (Cytosine-5-)-Methyltransferases/metabolism , Enzyme Inhibitors/therapeutic use , Epigenesis, Genetic/drug effects , Female , Femur/diagnostic imaging , Femur/pathology , Gene Expression Regulation, Neoplastic/drug effects , Histocompatibility Antigens/metabolism , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Histone-Lysine N-Methyltransferase/metabolism , Humans , Male , Mesenchymal Stem Cells/pathology , Mice , Middle Aged , Multiple Myeloma/complications , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Osteogenesis/drug effects , Osteogenesis/genetics , Xenograft Model Antitumor Assays
15.
Genome Med ; 12(1): 81, 2020 09 25.
Article in English | MEDLINE | ID: mdl-32977850

ABSTRACT

BACKGROUND: Systemic sclerosis (SSc) is a genetically complex autoimmune disease mediated by the interplay between genetic and epigenetic factors in a multitude of immune cells, with CD4+ T lymphocytes as one of the principle drivers of pathogenesis. METHODS: DNA samples exacted from CD4+ T cells of 48 SSc patients and 16 healthy controls were hybridized on MethylationEPIC BeadChip array. In parallel, gene expression was interrogated by hybridizing total RNA on Clariom™ S array. Downstream bioinformatics analyses were performed to identify correlating differentially methylated CpG positions (DMPs) and differentially expressed genes (DEGs), which were then confirmed utilizing previously published promoter capture Hi-C (PCHi-C) data. RESULTS: We identified 9112 and 3929 DMPs and DEGs, respectively. These DMPs and DEGs are enriched in functional categories related to inflammation and T cell biology. Furthermore, correlation analysis identified 17,500 possible DMP-DEG interaction pairs within a window of 5 Mb, and utilizing PCHi-C data, we observed that 212 CD4+ T cell-specific pairs of DMP-DEG also formed part of three-dimensional promoter-enhancer networks, potentially involving CTCF. Finally, combining PCHi-C data with SSc GWAS data, we identified four important SSc-associated susceptibility loci, TNIP1 (rs3792783), GSDMB (rs9303277), IL12RB1 (rs2305743), and CSK (rs1378942), that could potentially interact with DMP-DEG pairs cg17239269-ANXA6, cg19458020-CCR7, cg10808810-JUND, and cg11062629-ULK3, respectively. CONCLUSION: Our study unveils a potential link between genetic, epigenetic, and transcriptional deregulation in CD4+ T cells of SSc patients, providing a novel integrated view of molecular components driving SSc pathogenesis.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Epigenome , Genetic Loci , Genetic Predisposition to Disease , Scleroderma, Systemic/etiology , Transcriptome , CD4-Positive T-Lymphocytes/immunology , Computational Biology/methods , DNA Methylation , Epigenesis, Genetic , Epigenomics/methods , Gene Expression Profiling/methods , Genetic Association Studies , Humans , NF-kappa B/metabolism , Scleroderma, Systemic/metabolism , Signal Transduction
16.
Nucleic Acids Res ; 48(2): 665-681, 2020 01 24.
Article in English | MEDLINE | ID: mdl-31799621

ABSTRACT

Sirtuins 1 and 2 (SIRT1/2) are two NAD-dependent deacetylases with major roles in inflammation. In addition to deacetylating histones and other proteins, SIRT1/2-mediated regulation is coupled with other epigenetic enzymes. Here, we investigate the links between SIRT1/2 activity and DNA methylation in macrophage differentiation due to their relevance in myeloid cells. SIRT1/2 display drastic upregulation during macrophage differentiation and their inhibition impacts the expression of many inflammation-related genes. In this context, SIRT1/2 inhibition abrogates DNA methylation gains, but does not affect demethylation. Inhibition of hypermethylation occurs at many inflammatory loci, which results in more drastic upregulation of their expression upon macrophage polarization following bacterial lipopolysaccharide (LPS) challenge. SIRT1/2-mediated gains of methylation concur with decreases in activating histone marks, and their inhibition revert these histone marks to resemble an open chromatin. Remarkably, specific inhibition of DNA methyltransferases is sufficient to upregulate inflammatory genes that are maintained in a silent state by SIRT1/2. Both SIRT1 and SIRT2 directly interact with DNMT3B, and their binding to proinflammatory genes is lost upon exposure to LPS or through pharmacological inhibition of their activity. In all, we describe a novel role for SIRT1/2 to restrict premature activation of proinflammatory genes.


Subject(s)
DNA Methylation/genetics , Inflammation/genetics , Sirtuin 1/genetics , Sirtuin 2/genetics , Acetylation , Cell Differentiation/genetics , Chromatin/genetics , Gene Expression Regulation/genetics , Histones/genetics , Humans , Inflammation/chemically induced , Inflammation/pathology , Lipopolysaccharides/toxicity , Macrophages/metabolism , Promoter Regions, Genetic , Transcriptional Activation/genetics
17.
Genome Med ; 11(1): 66, 2019 10 29.
Article in English | MEDLINE | ID: mdl-31665078

ABSTRACT

BACKGROUND: Sepsis, a life-threatening organ dysfunction caused by a dysregulated systemic immune response to infection, associates with reduced responsiveness to subsequent infections. How such tolerance is acquired is not well understood but is known to involve epigenetic and transcriptional dysregulation. METHODS: Bead arrays were used to compare global DNA methylation changes in patients with sepsis, non-infectious systemic inflammatory response syndrome, and healthy controls. Bioinformatic analyses were performed to dissect functional reprogramming and signaling pathways related to the acquisition of these specific DNA methylation alterations. Finally, in vitro experiments using human monocytes were performed to test the induction of similar DNA methylation reprogramming. RESULTS: Here, we focused on DNA methylation changes associated with sepsis, given their potential role in stabilizing altered phenotypes. Tolerized monocytes from patients with sepsis display changes in their DNA methylomes with respect to those from healthy controls, affecting critical monocyte-related genes. DNA methylation profiles correlate with IL-10 and IL-6 levels, significantly increased in monocytes in sepsis, as well as with the Sequential Organ Failure Assessment score; the observed changes associate with TFs and pathways downstream to toll-like receptors and inflammatory cytokines. In fact, in vitro stimulation of toll-like receptors in monocytes results in similar gains and losses of methylation together with the acquisition of tolerance. CONCLUSION: We have identified a DNA methylation signature associated with sepsis that is downstream to the response of monocytes to inflammatory signals associated with the acquisition of a tolerized phenotype and organic dysfunction.


Subject(s)
Cytokines/genetics , DNA Methylation , DNA/analysis , Inflammation Mediators/metabolism , Inflammation/complications , Monocytes/pathology , Multiple Organ Failure/complications , Sepsis/diagnosis , Aged , Case-Control Studies , DNA/genetics , Female , Humans , Inflammation/genetics , Male , Middle Aged , Monocytes/immunology , Monocytes/metabolism , Multiple Organ Failure/genetics , Phenotype , Sepsis/etiology , Sepsis/metabolism , Signal Transduction
18.
Ann Rheum Dis ; 78(11): 1505-1516, 2019 11.
Article in English | MEDLINE | ID: mdl-31371305

ABSTRACT

OBJECTIVE: Rheumatoid arthritis (RA) is a chronic systemic autoimmune disease that mainly targets joints. Monocytes and macrophages are critical in RA pathogenesis and contribute to inflammatory lesions. These extremely plastic cells respond to extracellular signals which cause epigenomic changes that define their pathogenic phenotype. Here, we interrogated how DNA methylation alterations in RA monocytes are determined by extracellular signals. METHODS: High-throughput DNA methylation analyses of patients with RA and controls and in vitro cytokine stimulation were used to investigate the underlying mechanisms behind DNA methylation alterations in RA as well as their relationship with clinical parameters, including RA disease activity. RESULTS: The DNA methylomes of peripheral blood monocytes displayed significant changes and increased variability in patients with RA with respect to healthy controls. Changes in the monocyte methylome correlate with DAS28, in which high-activity patients are divergent from healthy controls in contrast to remission patients whose methylome is virtually identical to healthy controls. Indeed, the notion of a changing monocyte methylome is supported after comparing the profiles of same individuals at different stages of activity. We show how these changes are mediated by an increase in disease activity-associated cytokines, such as tumour necrosis factor alpha and interferons, as they recapitulate the DNA methylation changes observed in patients in vitro. CONCLUSION: We demonstrate a direct link between RA disease activity and the monocyte methylome through the action of inflammation-associated cytokines. Finally, we have obtained a DNA methylation-based mathematical formula that predicts inflammation-mediated disease activity for RA and other chronic immune-mediated inflammatory diseases.


Subject(s)
Arthritis, Rheumatoid/blood , Arthritis, Rheumatoid/genetics , Cytokines/blood , Epigenome/immunology , Inflammation Mediators/blood , Biomarkers/blood , DNA Methylation/immunology , Humans , Leukocytes, Mononuclear/immunology , Macrophages/immunology , Tumor Necrosis Factor-alpha/blood
19.
Sci Rep ; 7(1): 7594, 2017 08 08.
Article in English | MEDLINE | ID: mdl-28790320

ABSTRACT

Activation-induced cytidine deaminase (AID) triggers antibody diversification in B cells by catalysing deamination and subsequently mutating immunoglobulin (Ig) genes. Association of AID with RNA Pol II and occurrence of epigenetic changes during Ig gene diversification suggest participation of AID in epigenetic regulation. AID is mutated in hyper-IgM type 2 (HIGM2) syndrome. Here, we investigated the potential role of AID in the acquisition of epigenetic changes. We discovered that AID binding to the IgH locus promotes an increase in H4K20me3. In 293F cells, we demonstrate interaction between co-transfected AID and the three SUV4-20 histone H4K20 methyltransferases, and that SUV4-20H1.2, bound to the IgH switch (S) mu site, is replaced by SUV4-20H2 upon AID binding. Analysis of HIGM2 mutants shows that the AID truncated form W68X is impaired to interact with SUV4-20H1.2 and SUV4-20H2 and is unable to bind and target H4K20me3 to the Smu site. We finally show in mouse primary B cells undergoing class-switch recombination (CSR) that AID deficiency associates with decreased H4K20me3 levels at the Smu site. Our results provide a novel link between SUV4-20 enzymes and CSR and offer a new aspect of the interplay between AID and histone modifications in setting the epigenetic status of CSR sites.


Subject(s)
Cytidine Deaminase/genetics , Epigenesis, Genetic/immunology , Histone-Lysine N-Methyltransferase/genetics , Histones/genetics , Hyper-IgM Immunodeficiency Syndrome/genetics , Immunoglobulin Class Switching/genetics , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Binding Sites , Cell Line, Tumor , Cytidine Deaminase/immunology , Gene Expression Regulation , HEK293 Cells , HeLa Cells , Histone-Lysine N-Methyltransferase/immunology , Histones/immunology , Humans , Hyper-IgM Immunodeficiency Syndrome/immunology , Hyper-IgM Immunodeficiency Syndrome/pathology , Immunoglobulin G/genetics , Lipopolysaccharides/pharmacology , Lymphocyte Activation/drug effects , Mice , Mice, Inbred C57BL , Models, Biological , Mutation , Protein Binding , RNA Polymerase II/genetics , RNA Polymerase II/immunology , Signal Transduction
20.
Nat Commun ; 6: 7335, 2015 Jun 17.
Article in English | MEDLINE | ID: mdl-26081581

ABSTRACT

Common variable immunodeficiency (CVID), the most frequent primary immunodeficiency characterized by loss of B-cell function, depends partly on genetic defects, and epigenetic changes are thought to contribute to its aetiology. Here we perform a high-throughput DNA methylation analysis of this disorder using a pair of CVID-discordant MZ twins and show predominant gain of DNA methylation in CVID B cells with respect to those from the healthy sibling in critical B lymphocyte genes, such as PIK3CD, BCL2L1, RPS6KB2, TCF3 and KCNN4. Individual analysis confirms hypermethylation of these genes. Analysis in naive, unswitched and switched memory B cells in a CVID patient cohort shows impaired ability to demethylate and upregulate these genes in transitioning from naive to memory cells in CVID. Our results not only indicate a role for epigenetic alterations in CVID but also identify relevant DNA methylation changes in B cells that could explain the clinical manifestations of CVID individuals.


Subject(s)
B-Lymphocytes/metabolism , Common Variable Immunodeficiency/metabolism , DNA Methylation , Immunologic Memory , Case-Control Studies , Gene Expression Regulation , Humans , Twins, Monozygotic
SELECTION OF CITATIONS
SEARCH DETAIL
...