Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Blood Adv ; 8(11): 2880-2889, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38593222

ABSTRACT

ABSTRACT: Inhibitor development is the most severe complication of hemophilia A (HA) care and is associated with increased morbidity and mortality. This study aimed to use a novel immunoglobulin G epitope mapping method to explore the factor VIII (FVIII)-specific epitope profile in the SIPPET cohort population and to develop an epitope mapping-based inhibitor prediction model. The population consisted of 122 previously untreated patients with severe HA who were followed up for 50 days of exposure to FVIII or 3 years, whichever occurred first. Sampling was performed before FVIII treatment and at the end of the follow-up. The outcome was inhibitor development. The FVIII epitope repertoire was assessed by means of a novel random peptide phage-display assay. A least absolute shrinkage and selection operator (LASSO) regression model and a random forest model were fitted on posttreatment sample data and validated in pretreatment sample data. The predictive performance of these models was assessed by the C-statistic and a calibration plot. We identified 27 775 peptides putatively directed against FVIII, which were used as input for the statistical models. The C-statistic of the LASSO and random forest models were good at 0.78 (95% confidence interval [CI], 0.69-0.86) and 0.80 (95% CI, 0.72-0.89). Model calibration of both models was moderately good. Two statistical models, developed on data from a novel random peptide phage display assay, were used to predict inhibitor development before exposure to exogenous FVIII. These models can be used to set up diagnostic tests that predict the risk of inhibitor development before starting treatment with FVIII.


Subject(s)
Factor VIII , Hemophilia A , Peptide Library , Humans , Factor VIII/immunology , Epitope Mapping , Male , Cohort Studies , Epitopes/immunology
3.
Nat Immunol ; 23(7): 1076-1085, 2022 07.
Article in English | MEDLINE | ID: mdl-35761085

ABSTRACT

Memory B cells persist for a lifetime and rapidly differentiate into antibody-producing plasmablasts and plasma cells upon antigen re-encounter. The clonal relationship and evolution of memory B cells and circulating plasmablasts is not well understood. Using single-cell sequencing combined with isolation of specific antibodies, we found that in two healthy donors, the memory B cell repertoire was dominated by large IgM, IgA and IgG2 clonal families, whereas IgG1 families, including those specific for recall antigens, were of small size. Analysis of multiyear samples demonstrated stability of memory B cell clonal families and revealed that a large fraction of recently generated plasmablasts was derived from long-term memory B cell families and was found recurrently. Collectively, this study provides a systematic description of the structure, stability and dynamics of the human memory B cell pool and suggests that memory B cells may be active at any time point in the generation of plasmablasts.


Subject(s)
Memory B Cells , Plasma Cells , B-Lymphocytes , Cells, Cultured , Humans , Immunoglobulin G , Immunologic Memory
4.
Hematol Oncol ; 40(4): 541-553, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35451108

ABSTRACT

The spatial architecture of the lymphoid tissue in follicular lymphoma (FL) presents unique challenges to studying its immune microenvironment. We investigated the spatial interplay of T cells, macrophages, myeloid cells and natural killer T cells using multispectral immunofluorescence images of diagnostic biopsies of 32 patients. A deep learning-based image analysis pipeline was tailored to the needs of follicular lymphoma spatial histology research, enabling the identification of different immune cells within and outside neoplastic follicles. We analyzed the density and spatial co-localization of immune cells in the inter-follicular and intra-follicular regions of follicular lymphoma. Low inter-follicular density of CD8+FOXP3+ cells and co-localization of CD8+FOXP3+ with CD4+CD8+ cells were significantly associated with relapse (p = 0.0057 and p = 0.0019, respectively) and shorter time to progression after first-line treatment (Logrank p = 0.0097 and log-rank p = 0.0093, respectively). A low inter-follicular density of CD8+FOXP3+ cells is associated with increased risk of relapse independent of follicular lymphoma international prognostic index (FLIPI) (p = 0.038, Hazard ratio (HR) = 0.42 [0.19, 0.95], but not independent of co-localization of CD8+FOXP3+ with CD4+CD8+ cells (p = 0.43). Co-localization of CD8+FOXP3+ with CD4+CD8+ cells is predictors of time to relapse independent of the FLIPI score and density of CD8+FOXP3+ cells (p = 0.027, HR = 0.0019 [7.19 × 10-6 , 0.49], This suggests a potential role of inter-follicular CD8+FOXP3+ and CD4+CD8+ cells in the disease progression of FL, warranting further validation on larger patient cohorts.


Subject(s)
Lymphoma, Follicular , CD8-Positive T-Lymphocytes , Forkhead Transcription Factors , Humans , Lymphoma, Follicular/pathology , Neoplasm Recurrence, Local , Prognosis , Tumor Microenvironment
6.
Sci Rep ; 11(1): 18043, 2021 09 10.
Article in English | MEDLINE | ID: mdl-34508139

ABSTRACT

Interferons (IFNs) are key cytokines involved in alerting the immune system to viral infection. After IFN stimulation, cellular transcriptional profile critically changes, leading to the expression of several IFN stimulated genes (ISGs) that exert a wide variety of antiviral activities. Despite many ISGs have been already identified, a comprehensive network of coding and non-coding genes with a central role in IFN-response still needs to be elucidated. We performed a global RNA-Seq transcriptome profile of the HCV permissive human hepatoma cell line Huh7.5 and its parental cell line Huh7, upon IFN treatment, to define a network of genes whose coordinated modulation plays a central role in IFN-response. Our study adds molecular actors, coding and non-coding genes, to the complex molecular network underlying IFN-response and shows how systems biology approaches, such as correlation networks, network's topology and gene ontology analyses can be leveraged to this aim.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation , Interferon Regulatory Factors/genetics , Interferons/metabolism , Systems Biology/methods , Transcriptome , Binding Sites , Cell Line, Tumor , Computational Biology/methods , Gene Expression Profiling/methods , Gene Expression Regulation/drug effects , Gene Regulatory Networks , Humans , Interferon Regulatory Factors/metabolism , Interferons/pharmacology , Liver Neoplasms , Nucleotide Motifs , Protein Binding
7.
Hum Mol Genet ; 30(13): 1175-1187, 2021 06 17.
Article in English | MEDLINE | ID: mdl-33601422

ABSTRACT

Synaptic dysfunction and cognitive decline in Huntington's disease (HD) involve hyperactive A disintegrin and metalloproteinase domain-containing protein 10 (ADAM10). To identify the molecular mechanisms through which ADAM10 is associated with synaptic dysfunction in HD, we performed an immunoaffinity purification-mass spectrometry (IP-MS) study of endogenous ADAM10 in the brains of wild-type and HD mice. We found that proteins implicated in synapse organization, synaptic plasticity, and vesicle and organelles trafficking interact with ADAM10, suggesting that it may act as hub protein at the excitatory synapse. Importantly, the ADAM10 interactome is enriched in presynaptic proteins and ADAM10 co-immunoprecipitates with piccolo (PCLO), a key player in the recycling and maintenance of synaptic vesicles. In contrast, reduced ADAM10/PCLO immunoprecipitation occurs in the HD brain, with decreased density of synaptic vesicles in the reserve and docked pools at the HD presynaptic terminal. Conditional heterozygous deletion of ADAM10 in the forebrain of HD mice reduces active ADAM10 to wild-type level and normalizes ADAM10/PCLO complex formation and synaptic vesicle density and distribution. The results indicate that presynaptic ADAM10 and PCLO are a relevant component of HD pathogenesis.


Subject(s)
ADAM10 Protein/metabolism , Cytoskeletal Proteins/metabolism , Huntington Disease/metabolism , Neuropeptides/metabolism , Synaptic Vesicles/metabolism , ADAM10 Protein/genetics , Animals , Blotting, Western , Brain/metabolism , Brain/pathology , Brain/ultrastructure , Humans , Huntington Disease/genetics , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission , Presynaptic Terminals/metabolism , Protein Binding , Protein Interaction Maps/genetics , Proteomics/methods , Synaptic Vesicles/ultrastructure , Synaptosomes/metabolism , Tandem Mass Spectrometry/methods
8.
Neurobiol Dis ; 146: 105140, 2020 12.
Article in English | MEDLINE | ID: mdl-33065279

ABSTRACT

RUES2 cell lines represent the first collection of isogenic human embryonic stem cells (hESCs) carrying different pathological CAG lengths in the HTT gene. However, their neuronal differentiation potential has yet to be thoroughly evaluated. Here, we report that RUES2 during ventral telencephalic differentiation is biased towards medial ganglionic eminence (MGE). We also show that HD-RUES2 cells exhibit an altered MGE transcriptional signature in addition to recapitulating known HD phenotypes, with reduced expression of the neurodevelopmental regulators NEUROD1 and BDNF and increased cleavage of synaptically enriched N-cadherin. Finally, we identified the transcription factor SP1 as a common potential detrimental co-partner of muHTT by de novo motif discovery analysis on the LGE, MGE, and cortical genes differentially expressed in HD human pluripotent stem cells in our and additional datasets. Taken together, these observations suggest a broad deleterious effect of muHTT in the early phases of neuronal development that may unfold through its altered interaction with SP1.


Subject(s)
Biomarkers, Tumor/metabolism , Cell Differentiation/physiology , Human Embryonic Stem Cells/metabolism , Pluripotent Stem Cells/cytology , Receptors, Immunologic/metabolism , Cell Differentiation/drug effects , Human Embryonic Stem Cells/pathology , Humans , Huntington Disease/genetics , Huntington Disease/metabolism , Neurogenesis/physiology , Neurons/metabolism
9.
Cancer Res ; 80(21): 4693-4706, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32963011

ABSTRACT

Multiple myeloma is a plasma cell neoplasm characterized by the production of unfolded immunoglobulins, which cause endoplasmic reticulum (ER) stress and sensitivity to proteasome inhibition. The genomic landscape of multiple myeloma is characterized by the loss of several genes rarely mutated in other cancers that may underline specific weaknesses of multiple myeloma cells. One of these is FAM46C that is lost in more than 10% of patients with multiple myeloma. We show here that FAM46C is part of a new complex containing the ER-associated protein FNDC3A, which regulates trafficking and secretion and, by impairing autophagy, exacerbates proteostatic stress. Reconstitution of FAM46C in multiple myeloma cells that had lost it induced apoptosis and ER stress. Apoptosis was preceded by an increase of intracellular aggregates, which was not linked to increased translation of IgG mRNA, but rather to impairment of autophagy. Biochemical analysis showed that FAM46C requires interaction with ER bound protein FNDC3A to reside in the cytoplasmic side of the ER. FNDC3A was lost in some multiple myeloma cell lines. Importantly, depletion of FNDC3A increased the fitness of FAM46C-expressing cells and expression of FNDC3A in cells that had lost it recapitulated the effects of FAM46C, inducing aggregates and apoptosis. FAM46C and FNDC3A formed a complex that modulates secretion routes, increasing lysosome exocytosis. The cellular landscape generated by FAM46C/FNDC3A expression predicted sensitivity to sphingosine kinase inhibition. These results suggest that multiple myeloma cells remodel their trafficking machinery to cope with ER stress. SIGNIFICANCE: This study identifies a new multiple myeloma-specific tumor suppressor complex that regulates autophagy and unconventional secretion, highlighting the sensitivity of multiple myeloma cells to the accumulation of protein aggregates.


Subject(s)
Fibronectins/metabolism , Multiple Myeloma/pathology , Nucleotidyltransferases/metabolism , Protein Aggregation, Pathological/metabolism , Animals , Autophagy/physiology , Genes, Tumor Suppressor , Heterografts , Humans , Mice , Multiple Myeloma/genetics , Multiple Myeloma/metabolism , Nucleotidyltransferases/genetics , Protein Aggregates/physiology , Protein Transport/physiology , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
10.
Nucleic Acids Res ; 47(10): 5325-5340, 2019 06 04.
Article in English | MEDLINE | ID: mdl-30937446

ABSTRACT

Stem cell identity and plasticity are controlled by master regulatory genes and complex circuits also involving non-coding RNAs. Circular RNAs (circRNAs) are a class of RNAs generated from protein-coding genes by backsplicing, resulting in stable RNA structures devoid of free 5' and 3' ends. Little is known of the mechanisms of action of circRNAs, let alone in stem cell biology. In this study, for the first time, we determined that a circRNA controls mesenchymal stem cell (MSC) identity and differentiation. High-throughput MSC expression profiling from different tissues revealed a large number of expressed circRNAs. Among those, circFOXP1 was enriched in MSCs compared to differentiated mesodermal derivatives. Silencing of circFOXP1 dramatically impaired MSC differentiation in culture and in vivo. Furthermore, we demonstrated a direct interaction between circFOXP1 and miR-17-3p/miR-127-5p, which results in the modulation of non-canonical Wnt and EGFR pathways. Finally, we addressed the interplay between canonical and non-canonical Wnt pathways. Reprogramming to pluripotency of MSCs reduced circFOXP1 and non-canonical Wnt, whereas canonical Wnt was boosted. The opposing effect was observed during generation of MSCs from human pluripotent stem cells. Our results provide unprecedented evidence for a regulatory role for circFOXP1 as a gatekeeper of pivotal stem cell molecular networks.


Subject(s)
Forkhead Transcription Factors/metabolism , MicroRNAs/metabolism , RNA , Repressor Proteins/metabolism , Cell Differentiation , Cell Nucleus/metabolism , Cell Proliferation , Cytoplasm/metabolism , ErbB Receptors/metabolism , Exoribonucleases/metabolism , Fibroblasts/metabolism , Gene Expression Profiling , Gene Silencing , HEK293 Cells , Humans , Immunophenotyping , Mesenchymal Stem Cells/cytology , Mesoderm/metabolism , Oligonucleotide Array Sequence Analysis , Pluripotent Stem Cells/cytology , RNA, Circular , RNA, Small Interfering/metabolism , Sequence Analysis, RNA , Stem Cells/cytology , Wnt Proteins/metabolism
11.
Methods Mol Biol ; 1959: 247-259, 2019.
Article in English | MEDLINE | ID: mdl-30852827

ABSTRACT

The diagnostic accuracy of biomarker-based approaches can be considerably improved by combining multiple markers. A biomarker's capacity to identify specific subjects is usually assessed using receiver operating characteristic (ROC) curves. Multimarker signatures are complicated to select as data signatures must be integrated using sophisticated statistical methods. CombiROC, developed as a user-friendly web tool, helps researchers to accurately determine optimal combinations of markers identified by a range of omics methods. With CombiROC, data of different types, such as proteomics and transcriptomics, can be analyzed using Sensitivity/Specificity filters: the number of candidate marker panels arising from combinatorial analysis is easily optimized bypassing limitations imposed by the nature of different experimental approaches. Users have full control over initial selection stringency, then CombiROC computes sensitivity and specificity for all marker combinations, determines performance for the best combinations, and produces ROC curves for automatic comparisons. All steps can be visualized in a graphic interface. CombiROC is designed without hard-coded thresholds, to allow customized fitting of each specific dataset: this approach dramatically reduces computational burden and false-negative rates compared to fixed thresholds. CombiROC can be accessed at www.combiroc.eu .


Subject(s)
Biomarkers , Computational Biology/methods , Gene Expression Profiling , Proteomics , Software , Data Interpretation, Statistical , Gene Expression Profiling/methods , Humans , Proteome , Proteomics/methods , ROC Curve , Transcriptome , User-Computer Interface
12.
J Exp Med ; 216(2): 317-336, 2019 02 04.
Article in English | MEDLINE | ID: mdl-30655308

ABSTRACT

Altered control of T follicular helper (Tfh) cells can lead to generation of autoantibodies and autoimmune manifestations. Signaling pathways that selectively limit pathogenic responses without affecting the protective function of Tfh cells are unknown. Here we show that the ATP-gated ionotropic P2X7 receptor restricts the expansion of aberrant Tfh cells and the generation of self-reactive antibodies in experimental murine lupus, but its activity is dispensable for the expansion of antigen-specific Tfh cells during vaccination. P2X7 stimulation promotes caspase-mediated pyroptosis of Tfh cells and controls the development of pathogenic ICOS+ IFN-γ-secreting cells. Circulating Tfh cells from patients with systemic lupus erythematosus (SLE) but not primary antiphospholipid syndrome (PAPS), a nonlupus systemic autoimmune disease, were hyporesponsive to P2X7 stimulation and resistant to P2X7-mediated inhibition of cytokine-driven expansion. These data point to the P2X7 receptor as a checkpoint regulator of Tfh cells; thus, restoring P2X7 activity in SLE patients could selectively limit the progressive amplification of pathogenic autoantibodies, which deteriorate patients' conditions.


Subject(s)
Lupus Erythematosus, Systemic/immunology , Receptors, Purinergic P2X7/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Autoantibodies/genetics , Autoantibodies/immunology , Disease Models, Animal , Interferon-gamma/genetics , Interferon-gamma/immunology , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/pathology , Mice , Mice, Knockout , Pyroptosis/genetics , Pyroptosis/immunology , Receptors, Purinergic P2X7/genetics , T-Lymphocytes, Helper-Inducer/pathology
13.
Eur J Immunol ; 49(1): 96-111, 2019 01.
Article in English | MEDLINE | ID: mdl-30431161

ABSTRACT

Whether human IL-10-producing regulatory T cells ("Tr1") represent a distinct differentiation lineage or an unstable activation stage remains a key unsolved issue. Here, we report that Eomesodermin (Eomes) acted as a lineage-defining transcription factor in human IFN-γ/IL-10 coproducing Tr1-like cells. In vivo occurring Tr1-like cells expressed Eomes, and were clearly distinct from all other CD4+ T-cell subsets, including conventional cytotoxic CD4+ T cells. They expressed Granzyme (Gzm) K, but had lost CD40L and IL-7R expression. Eomes antagonized the Th17 fate, and directly controlled IFN-γ and GzmK expression. However, Eomes binding to the IL-10 promoter was not detectable in human CD4+ T cells, presumably because critical Tbox binding sites of the mouse were not conserved. A precommitment to a Tr1-like fate, i.e. concominant induction of Eomes, GzmK, and IFN-γ, was promoted by IL-4 and IL-12-secreting myeloid dendritic cells. Consistently, Th1 effector memory cells contained precommitted Eomes+ GzmK+ T cells. Stimulation with T-cell receptor (TCR) agonists and IL-27 promoted the generation of Tr1-like effector cells by inducing switching from CD40L to IL-10. Importantly, CD4+ Eomes+ T-cell subsets were present in lymphoid and nonlymphoid tissues, and their frequencies varied systemically in patients with inflammatory bowel disease and graft-versus-host disease. We propose that Eomes+ Tr1-like cells are effector cells of a unique GzmK-expressing CD4+ T-cell subset.


Subject(s)
Graft vs Host Disease/immunology , Inflammatory Bowel Diseases/immunology , T-Box Domain Proteins/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology , Animals , Cell Differentiation , Cell Lineage , Cells, Cultured , Gene Expression Regulation , Granzymes/metabolism , Humans , Immunologic Memory , Interferon-gamma/metabolism , Interleukin-10/metabolism , Mice , T-Box Domain Proteins/genetics
14.
Cereb Cortex ; 28(5): 1846-1856, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29351579

ABSTRACT

During differentiation, neurons progressively restrict their fate repressing the expression of specific genes. Here we describe the involvement in such developmental steps of the methyl-CpG binding protein 2 (MeCP2), an epigenetic factor that participates to chromatin folding and transcriptional regulation. We previously reported that, due to transcriptional impairments, the maturation of Mecp2 null neurons is delayed. To evaluate whether this could stem from altered progenitors proliferation and differentiation, we investigated whether lack of Mecp2 affects these features both in vitro and in vivo. We show that in Mecp2 null embryonic cortexes the expression of genes defining the identity of proliferating neuroprogenitors is enriched and that their permanence in the G1 phase is prolonged. Moreover, the number of cells transitioning from a stage of maturation to a more mature one is increased in Mecp2 null embryonic cortices, in line with the central role of G1 for cell identity refinement. We thus suggest that, possibly due to the lack of proper transcriptional control normally exerted by Mecp2, fate refinement is impaired in developing null cells. We propose that the maturation delay affecting the developing Mecp2 null cortex originates, at least in part, from deranged mechanisms of cell fate refinement.


Subject(s)
Cerebral Cortex/cytology , Cerebral Cortex/embryology , Gene Expression Regulation, Developmental/genetics , Methyl-CpG-Binding Protein 2/deficiency , Neurons/pathology , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Bromodeoxyuridine , Cells, Cultured , Cyclin D1/metabolism , Deoxyuridine/analogs & derivatives , Deoxyuridine/toxicity , Embryo, Mammalian/cytology , Embryo, Mammalian/drug effects , Epidermal Growth Factor/pharmacology , Female , Fibroblast Growth Factors/pharmacology , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Methyl-CpG-Binding Protein 2/genetics , Mice , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Neurogenesis/drug effects , Neurogenesis/genetics , Pregnancy , Proliferating Cell Nuclear Antigen/metabolism , RNA, Messenger
15.
Sci Rep ; 7: 45477, 2017 03 30.
Article in English | MEDLINE | ID: mdl-28358118

ABSTRACT

Diagnostic accuracy can be improved considerably by combining multiple markers, whose performance in identifying diseased subjects is usually assessed via receiver operating characteristic (ROC) curves. The selection of multimarker signatures is a complicated process that requires integration of data signatures with sophisticated statistical methods. We developed a user-friendly tool, called CombiROC, to help researchers accurately determine optimal markers combinations from diverse omics methods. With CombiROC data from different domains, such as proteomics and transcriptomics, can be analyzed using sensitivity/specificity filters: the number of candidate marker panels rising from combinatorial analysis is easily optimized bypassing limitations imposed by the nature of different experimental approaches. Leaving to the user full control on initial selection stringency, CombiROC computes sensitivity and specificity for all markers combinations, performances of best combinations and ROC curves for automatic comparisons, all visualized in a graphic interface. CombiROC was designed without hard-coded thresholds, allowing a custom fit to each specific data: this dramatically reduces the computational burden and lowers the false negative rates given by fixed thresholds. The application was validated with published data, confirming the marker combination already originally described or even finding new ones. CombiROC is a novel tool for the scientific community freely available at http://CombiROC.eu.


Subject(s)
Biomarkers/analysis , User-Computer Interface , Area Under Curve , Biomarkers/blood , Genomics , Hepatitis, Autoimmune/diagnosis , Hepatitis, Autoimmune/metabolism , Humans , Internet , Lymphoma/diagnosis , Lymphoma/genetics , MicroRNAs/blood , Myotonic Dystrophy/diagnosis , Myotonic Dystrophy/genetics , Proteomics , ROC Curve , Sensitivity and Specificity
16.
J Biol Chem ; 292(7): 2903-2915, 2017 02 17.
Article in English | MEDLINE | ID: mdl-28077577

ABSTRACT

Upon T cell receptor stimulation, CD4+ T helper (Th) lymphocytes release extracellular vesicles (EVs) containing microRNAs. However, no data are available on whether human CD4+ T cell subsets release EVs containing different pattern of microRNAs. The present work aimed at filling this gap by assessing the microRNA content in EVs released upon in vitro T cell receptor stimulation of Th1, Th17, and T regulatory (Treg) cells. Our results indicate that EVs released by Treg cells are significantly different compared with those released by the other subsets. In particular, miR-146a-5p, miR-150-5p, and miR-21-5p are enriched, whereas miR-106a-5p, miR-155-5p, and miR-19a-3p are depleted in Treg-derived EVs. The in vitro identified EV-associated microRNA signature was increased in serum of autoimmune patients with psoriasis and returned to healthy levels upon effective treatment with etanercept, a biological drug targeting the TNF pathway and suppressing inflammation. Moreover, Gene Set Enrichment Analysis showed an over-representation of genes relevant for T cell activation, such as CD40L, IRAK1, IRAK2, STAT1, and c-Myb in the list of validated targets of Treg-derived EV miRNAs. At functional level, Treg-derived (but not Th1/Th17-derived) EVs inhibited CD4+ T cell proliferation and suppressed two relevant targets of miR-146a-5p: STAT1 and IRAK2. In conclusion, our work identified the miRNAs specifically released by different human CD4+ T cell subsets and started to unveil the potential use of their quantity in human serum to mark the pathological elicitation of these cells in vivo and their biological effect in cell to cell communication during the adaptive immune response.


Subject(s)
Autoimmune Diseases/genetics , CD4-Positive T-Lymphocytes/cytology , Extracellular Vesicles/metabolism , MicroRNAs/genetics , T-Lymphocyte Subsets , Autoimmune Diseases/immunology , CD4-Positive T-Lymphocytes/immunology , Cells, Cultured , Etanercept/therapeutic use , Humans , MicroRNAs/blood , Psoriasis/blood , Psoriasis/drug therapy , Psoriasis/genetics
17.
Nat Commun ; 7: 11903, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27301576

ABSTRACT

Stem cell identity depends on the integration of extrinsic and intrinsic signals, which directly influence the maintenance of their epigenetic state. Although Myc transcription factors play a major role in stem cell self-renewal and pluripotency, their integration with signalling pathways and epigenetic regulators remains poorly defined. We addressed this point by profiling the gene expression and epigenetic pattern in ESCs whose growth depends on conditional Myc activity. Here we show that Myc potentiates the Wnt/ß-catenin signalling pathway, which cooperates with the transcriptional regulatory network in sustaining ESC self-renewal. Myc activation results in the transcriptional repression of Wnt antagonists through the direct recruitment of PRC2 on these targets. The consequent potentiation of the autocrine Wnt/ß-catenin signalling induces the transcriptional activation of the endogenous Myc family members, which in turn activates a Myc-driven self-reinforcing circuit. Thus, our data unravel a Myc-dependent self-propagating epigenetic memory in the maintenance of ESC self-renewal capacity.


Subject(s)
Gene Regulatory Networks/genetics , Mouse Embryonic Stem Cells/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Animals , Cell Self Renewal/drug effects , Epigenesis, Genetic/drug effects , Feedback, Physiological/drug effects , Gene Regulatory Networks/drug effects , Leukemia Inhibitory Factor/pharmacology , Mice , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/drug effects , Polycomb-Group Proteins/metabolism , Transcription, Genetic/drug effects , Wnt Signaling Pathway/drug effects , Wnt Signaling Pathway/genetics
18.
Brief Bioinform ; 17(2): 204-12, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26238539

ABSTRACT

The high-throughput analysis of microRNAs (miRNAs) circulating within the blood of healthy and diseased individuals is an active area of biomarker research. Whereas quantitative real-time reverse transcription polymerase chain reaction (qPCR)-based methods are widely used, it is yet unresolved how the data should be normalized. Here, we show that a combination of different algorithms results in the identification of candidate reference miRNAs that can be exploited as normalizers, in both discovery and validation phases. Using the methodology considered here, we identify normalizers that are able to reduce nonbiological variation in the data and we present several case studies, to illustrate the relevance in the context of physiological or pathological scenarios. In conclusion, the discovery of stable reference miRNAs from high-throughput studies allows appropriate normalization of focused qPCR assays.


Subject(s)
Algorithms , Gene Expression Profiling/methods , High-Throughput Nucleotide Sequencing/methods , MicroRNAs/blood , MicroRNAs/genetics , Real-Time Polymerase Chain Reaction/methods , Biomarkers/blood , Gene Expression Profiling/standards , High-Throughput Nucleotide Sequencing/standards , Humans , MicroRNAs/standards , Real-Time Polymerase Chain Reaction/standards , Reference Values , Reproducibility of Results , Sensitivity and Specificity
19.
Nucleic Acids Res ; 43(W1): W487-92, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25897123

ABSTRACT

The miRBase is the official miRNA repository which keeps the annotation updated on newly discovered miRNAs: it is also used as a reference for the design of miRNA profiling platforms. Nomenclature ambiguities generated by loosely updated platforms and design errors lead to incompatibilities among platforms, even from the same vendor. Published miRNA lists are thus generated with different profiling platforms that refer to diverse and not updated annotations. This greatly compromises searches, comparisons and analyses that rely on miRNA names only without taking into account the mature sequences, which is particularly critic when such analyses are carried over automatically. In this paper we introduce miRiadne, a web tool to harmonize miRNA nomenclature, which takes into account the original miRBase versions from 10 up to 21, and annotations of 40 common profiling platforms from nine brands that we manually curated. miRiadne uses the miRNA mature sequence to link miRBase versions and/or platforms to prevent nomenclature ambiguities. miRiadne was designed to simplify and support biologists and bioinformaticians in re-annotating their own miRNA lists and/or data sets. As Ariadne helped Theseus in escaping the mythological maze, miRiadne will help the miRNA researcher in escaping the nomenclature maze. miRiadne is freely accessible from the URL http://www.miriadne.org.


Subject(s)
MicroRNAs/chemistry , Molecular Sequence Annotation , Software , Terminology as Topic , Internet , MicroRNAs/metabolism
20.
Neurosci Biobehav Rev ; 46 Pt 2: 187-201, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24594195

ABSTRACT

Rett syndrome (RTT) is a devastating genetic disorder that worldwide represents the most common genetic cause of severe intellectual disability in females. Most cases are caused by mutations in the X-linked MECP2 gene. Several recent studies have demonstrated that RTT mimicking animal models do not develop an irreversible condition and phenotypic rescue is possible. However, no cure for RTT has been identified so far, and patients are only given symptomatic and supportive treatments. The development of clinical applications imposes a more comprehensive knowledge of MeCP2 functional role(s) and their relevance for RTT pathobiology. Herein, we thoroughly survey the knowledge about MeCP2 structure and functions, highlighting the necessity of identifying more functional domains and the value of molecular genetics. Given that, in our opinion, RTT ultimately is generated by perturbations in gene transcription and so far no genes/pathways have been consistently linked to a dysfunctional MeCP2, we have used higher-level bioinformatic analyses to identify commonly deregulated mechanisms in MeCP2-defective samples. In this review we present our results and discuss the possible value of the utilized approach.


Subject(s)
Gene Expression Regulation, Developmental/genetics , Methyl-CpG-Binding Protein 2/genetics , Methyl-CpG-Binding Protein 2/metabolism , Rett Syndrome/genetics , Rett Syndrome/metabolism , Signal Transduction/genetics , Animals , Computational Biology , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...