Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Am J Hum Genet ; 110(10): 1648-1660, 2023 10 05.
Article in English | MEDLINE | ID: mdl-37673065

ABSTRACT

X-linked myotubular myopathy (XLMTM) is a severe congenital disease characterized by profound muscle weakness, respiratory failure, and early death. No approved therapy for XLMTM is currently available. Adeno-associated virus (AAV)-mediated gene replacement therapy has shown promise as an investigational therapeutic strategy. We aimed to characterize the transcriptomic changes in muscle biopsies of individuals with XLMTM who received resamirigene bilparvovec (AT132; rAAV8-Des-hMTM1) in the ASPIRO clinical trial and to identify potential biomarkers that correlate with therapeutic outcome. We leveraged RNA-sequencing data from the muscle biopsies of 15 study participants and applied differential expression analysis, gene co-expression analysis, and machine learning to characterize the transcriptomic changes at baseline (pre-dose) and at 24 and 48 weeks after resamirigene bilparvovec dosing. As expected, MTM1 expression levels were significantly increased after dosing (p < 0.0001). Differential expression analysis identified upregulated genes after dosing that were enriched in several pathways, including lipid metabolism and inflammatory response pathways, and downregulated genes were enriched in cell-cell adhesion and muscle development pathways. Genes involved in inflammatory and immune pathways were differentially expressed between participants exhibiting ventilator support reduction of either greater or less than 6 h/day after gene therapy compared to pre-dosing. Co-expression analysis identified similarly regulated genes, which were grouped into modules. Finally, the machine learning model identified five genes, including MTM1, as potential RNA biomarkers to monitor the progress of AAV gene replacement therapy. These findings further extend our understanding of AAV-mediated gene therapy in individuals with XLMTM at the transcriptomic level.


Subject(s)
Myopathies, Structural, Congenital , Transcriptome , Humans , Biomarkers/metabolism , Gene Expression Profiling , Genetic Therapy , Muscle, Skeletal/metabolism , Myopathies, Structural, Congenital/genetics , Myopathies, Structural, Congenital/therapy , Myopathies, Structural, Congenital/pathology , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , RNA/metabolism , Transcriptome/genetics
2.
Mol Ther Nucleic Acids ; 32: 229-246, 2023 Jun 13.
Article in English | MEDLINE | ID: mdl-37090420

ABSTRACT

Sickle cell disease (SCD) is due to a mutation in the ß-globin gene causing production of the toxic sickle hemoglobin (HbS; α2ßS 2). Transplantation of autologous hematopoietic stem and progenitor cells (HSPCs) transduced with lentiviral vectors (LVs) expressing an anti-sickling ß-globin (ßAS) is a promising treatment; however, it is only partially effective, and patients still present elevated HbS levels. Here, we developed a bifunctional LV expressing ßAS3-globin and an artificial microRNA (amiRNA) specifically downregulating ßS-globin expression with the aim of reducing HbS levels and favoring ßAS3 incorporation into Hb tetramers. Efficient transduction of SCD HSPCs by the bifunctional LV led to a substantial decrease of ßS-globin transcripts in HSPC-derived erythroid cells, a significant reduction of HbS+ red cells, and effective correction of the sickling phenotype, outperforming ßAS gene addition and BCL11A gene silencing strategies. The bifunctional LV showed a standard integration profile, and neither HSPC viability, engraftment, and multilineage differentiation nor the erythroid transcriptome and miRNAome were affected by the treatment, confirming the safety of this therapeutic strategy. In conclusion, the combination of gene addition and gene silencing strategies can improve the efficacy of current LV-based therapeutic approaches without increasing the mutagenic vector load, thus representing a novel treatment for SCD.

3.
Nat Commun ; 13(1): 6618, 2022 11 04.
Article in English | MEDLINE | ID: mdl-36333351

ABSTRACT

Sickle cell disease and ß-thalassemia affect the production of the adult ß-hemoglobin chain. The clinical severity is lessened by mutations that cause fetal γ-globin expression in adult life (i.e., the hereditary persistence of fetal hemoglobin). Mutations clustering ~200 nucleotides upstream of the HBG transcriptional start sites either reduce binding of the LRF repressor or recruit the KLF1 activator. Here, we use base editing to generate a variety of mutations in the -200 region of the HBG promoters, including potent combinations of four to eight γ-globin-inducing mutations. Editing of patient hematopoietic stem/progenitor cells is safe, leads to fetal hemoglobin reactivation and rescues the pathological phenotype. Creation of a KLF1 activator binding site is the most potent strategy - even in long-term repopulating hematopoietic stem/progenitor cells. Compared with a Cas9-nuclease approach, base editing avoids the generation of insertions, deletions and large genomic rearrangements and results in higher γ-globin levels. Our results demonstrate that base editing of HBG promoters is a safe, universal strategy for treating ß-hemoglobinopathies.


Subject(s)
Anemia, Sickle Cell , beta-Thalassemia , Humans , Fetal Hemoglobin/genetics , Fetal Hemoglobin/metabolism , gamma-Globins/genetics , beta-Thalassemia/genetics , beta-Thalassemia/therapy , Anemia, Sickle Cell/genetics , Hematopoietic Stem Cells/metabolism
4.
J Mol Biol ; 434(11): 167560, 2022 06 15.
Article in English | MEDLINE | ID: mdl-35662457

ABSTRACT

The advent of single-cell sequencing is providing unprecedented opportunities to disentangle tissue complexity and investigate cell identities and functions. However, the analysis of single cell data is a challenging, multi-step process that requires both advanced computational skills and biological sensibility. When dealing with single cell RNA-seq (scRNA-seq) data, the presence of technical artifacts, noise, and biological biases imposes to first identify, and eventually remove, unreliable signals from low-quality cells and unwanted sources of variation that might affect the efficacy of subsequent downstream modules. Pre-processing and quality control (QC) of scRNA-seq data is a laborious process consisting in the manual combination of different computational strategies to quantify QC-metrics and define optimal sets of pre-processing parameters. Here we present popsicleR, a R package to interactively guide skilled and unskilled command line-users in the pre-processing and QC analysis of scRNA-seq data. The package integrates, into several main wrapper functions, methods derived from widely used pipelines for the estimation of quality-control metrics, filtering of low-quality cells, data normalization, removal of technical and biological biases, and for cell clustering and annotation. popsicleR starts from either the output files of the Cell Ranger pipeline from 10X Genomics or from a feature-barcode matrix of raw counts generated from any scRNA-seq technology. Open-source code, installation instructions, and a case study tutorial are freely available at https://github.com/bicciatolab/popsicleR.


Subject(s)
RNA-Seq , Single-Cell Analysis , Software , Gene Expression Profiling/methods , Quality Control , RNA-Seq/methods , Single-Cell Analysis/methods
5.
Nat Commun ; 12(1): 2340, 2021 04 20.
Article in English | MEDLINE | ID: mdl-33879786

ABSTRACT

Cancer is characterized by pervasive epigenetic alterations with enhancer dysfunction orchestrating the aberrant cancer transcriptional programs and transcriptional dependencies. Here, we epigenetically characterize human colorectal cancer (CRC) using de novo chromatin state discovery on a library of different patient-derived organoids. By exploring this resource, we unveil a tumor-specific deregulated enhancerome that is cancer cell-intrinsic and independent of interpatient heterogeneity. We show that the transcriptional coactivators YAP/TAZ act as key regulators of the conserved CRC gained enhancers. The same YAP/TAZ-bound enhancers display active chromatin profiles across diverse human tumors, highlighting a pan-cancer epigenetic rewiring which at single-cell level distinguishes malignant from normal cell populations. YAP/TAZ inhibition in established tumor organoids causes extensive cell death unveiling their essential role in tumor maintenance. This work indicates a common layer of YAP/TAZ-fueled enhancer reprogramming that is key for the cancer cell state and can be exploited for the development of improved therapeutic avenues.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Colorectal Neoplasms/genetics , Enhancer Elements, Genetic , Epigenesis, Genetic , Trans-Activators/genetics , Transcription Factors/genetics , Gene Expression Regulation, Neoplastic , Histone Code , Humans , Models, Genetic , Organoids/metabolism , RNA-Seq , Single-Cell Analysis , Transcriptional Coactivator with PDZ-Binding Motif Proteins , Tumor Cells, Cultured , YAP-Signaling Proteins
6.
Nat Commun ; 12(1): 2237, 2021 04 14.
Article in English | MEDLINE | ID: mdl-33854047

ABSTRACT

Acquisition of cell-associated tumor antigens by type 1 dendritic cells (cDC1) is essential to induce and sustain tumor specific CD8+ T cells via cross-presentation. Here we show that capture and engulfment of cell associated antigens by tissue resident lung cDC1 is inhibited during progression of mouse lung tumors. Mechanistically, loss of phagocytosis is linked to tumor-mediated downregulation of the phosphatidylserine receptor TIM4, that is highly expressed in normal lung resident cDC1. TIM4 receptor blockade and conditional cDC1 deletion impair activation of tumor specific CD8+ T cells and promote tumor progression. In human lung adenocarcinomas, TIM4 transcripts increase the prognostic value of a cDC1 signature and predict responses to PD-1 treatment. Thus, TIM4 on lung resident cDC1 contributes to immune surveillance and its expression is suppressed in advanced tumors.


Subject(s)
Antigens, Neoplasm/immunology , Dendritic Cells/immunology , Lung Neoplasms/immunology , Membrane Proteins/immunology , Adenocarcinoma/genetics , Adenocarcinoma/immunology , Animals , Antigens, Neoplasm/genetics , CD8-Positive T-Lymphocytes/immunology , Cross-Priming , Humans , Immunologic Surveillance , Lung/immunology , Lung Neoplasms/genetics , Membrane Proteins/genetics , Mice
7.
Nat Cancer ; 2(2): 174-188, 2021 02.
Article in English | MEDLINE | ID: mdl-33644767

ABSTRACT

Glioblastoma (GBM) is a devastating human malignancy. GBM stem-like cells (GSCs) drive tumor initiation and progression. Yet, the molecular determinants defining GSCs in their native state in patients remain poorly understood. Here we used single cell datasets and identified GSCs at the apex of the differentiation hierarchy of GBM. By reconstructing the GSCs' regulatory network, we identified the YAP/TAZ coactivators as master regulators of this cell state, irrespectively of GBM subtypes. YAP/TAZ are required to install GSC properties in primary cells downstream of multiple oncogenic lesions, and required for tumor initiation and maintenance in vivo in different mouse and human GBM models. YAP/TAZ act as main roadblock of GSC differentiation and their inhibition irreversibly lock differentiated GBM cells into a non-tumorigenic state, preventing plasticity and regeneration of GSC-like cells. Thus, GSC identity is linked to a key molecular hub integrating genetics and microenvironmental inputs within the multifaceted biology of GBM.


Subject(s)
Brain Neoplasms , Glioblastoma , Animals , Brain Neoplasms/genetics , Carcinogenesis/pathology , Cell Plasticity , Glioblastoma/genetics , Humans , Mice , Neoplastic Stem Cells/pathology , Single-Cell Analysis
8.
NPJ Precis Oncol ; 5(1): 4, 2021 Feb 04.
Article in English | MEDLINE | ID: mdl-33542466

ABSTRACT

Disease progression of myeloproliferative neoplasms is the result of increased genomic complexity. Since the ability to predict disease evolution is crucial for clinical decisions, we studied single-cell genomics and transcriptomics of CD34-positive cells from a primary myelofibrosis (PMF) patient who progressed to acute myeloid leukemia (AML) while receiving Ruxolitinib. Single-cell genomics allowed the reconstruction of clonal hierarchy and demonstrated that TET2 was the first mutated gene while FLT3 was the last one. Disease evolution was accompanied by increased clonal heterogeneity and mutational rate, but clones carrying TP53 and FLT3 mutations were already present in the chronic phase. Single-cell transcriptomics unraveled repression of interferon signaling suggesting an immunosuppressive effect exerted by Ruxolitinib. Moreover, AML transformation was associated with a differentiative block and immune escape. These results suggest that single-cell analysis can unmask tumor heterogeneity and provide meaningful insights about PMF progression that might guide personalized therapy.

9.
Brief Bioinform ; 22(1): 20-29, 2021 01 18.
Article in English | MEDLINE | ID: mdl-32363378

ABSTRACT

Recent advances in single-cell technologies are providing exciting opportunities for dissecting tissue heterogeneity and investigating cell identity, fate and function. This is a pristine, exploding field that is flooding biologists with a new wave of data, each with its own specificities in terms of complexity and information content. The integrative analysis of genomic data, collected at different molecular layers from diverse cell populations, holds promise to address the full-scale complexity of biological systems. However, the combination of different single-cell genomic signals is computationally challenging, as these data are intrinsically heterogeneous for experimental, technical and biological reasons. Here, we describe the computational methods for the integrative analysis of single-cell genomic data, with a focus on the integration of single-cell RNA sequencing datasets and on the joint analysis of multimodal signals from individual cells.


Subject(s)
Computational Biology/methods , RNA-Seq/methods , Single-Cell Analysis/methods , Animals , Humans
10.
iScience ; 23(4): 101018, 2020 Apr 24.
Article in English | MEDLINE | ID: mdl-32283524

ABSTRACT

Erythroid commitment and differentiation are regulated by the coordinated action of a host of transcription factors, including GATA2 and GATA1. Here, we explored GATA-mediated transcriptional regulation through the integrative analysis of gene expression, chromatin modifications, and GATA factors' binding in human multipotent hematopoietic stem/progenitor cells, early erythroid progenitors, and late precursors. A progressive loss of H3K27 acetylation and a diminished usage of active enhancers and super-enhancers were observed during erythroid commitment and differentiation. GATA factors mediate transcriptional changes through a stage-specific interplay with regulatory elements: GATA1 binds different sets of regulatory elements in erythroid progenitors and precursors and controls the transcription of distinct genes during commitment and differentiation. Importantly, our results highlight a pivotal role of promoters in determining the transcriptional program activated upon erythroid differentiation. Finally, we demonstrated that GATA1 binding to a stage-specific super-enhancer sustains the expression of the KIT receptor in human erythroid progenitors.

11.
IUBMB Life ; 72(1): 10-26, 2020 01.
Article in English | MEDLINE | ID: mdl-31574210

ABSTRACT

The members of the GATA family of transcription factors have homologous zinc fingers and bind to similar sequence motifs. Recent advances in genome-wide technologies and the integration of bioinformatics data have led to a better understanding of how GATA factors regulate gene expression; GATA-factor-induced transcriptional and epigenetic changes have now been analyzed at unprecedented levels of detail. Here, we review the results of genome-wide studies of GATA factor occupancy in human and murine cell lines and primary cells (as determined by chromatin immunoprecipitation sequencing), and then discuss the molecular mechanisms underlying the mediation of transcriptional and epigenetic regulation by GATA factors.


Subject(s)
Epigenesis, Genetic , GATA Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic , Genome , Neoplasms/pathology , Animals , Computational Biology , GATA Transcription Factors/genetics , Humans , Neoplasms/genetics , Neoplasms/metabolism
12.
Cell Signal ; 59: 131-140, 2019 07.
Article in English | MEDLINE | ID: mdl-30926389

ABSTRACT

Elastic fibres undergo aberrant mineralization in genetic as well as in acquired pathologic conditions causing severe impairment of tissue mechanical properties. Despite the number of investigations performed so far, the pathogenesis of these alterations is still elusive, due to both the complexity of the elastin network and the involvement of many genes and/or pro-osteogenic signalling pathways. Whole Exome Sequencing (WES) was performed on DNA from three patients affected by beta-thalassemia exhibiting soft connective tissue calcification. WES data were analysed with a bioinformatic approach, allowing to screen and to select genes carrying rare sequence variants. These genes were matched with those present in Extracellular Matrix DB. This approach enables to shed light on the involvement of the extracellular matrix in the occurrence of ectopic calcification. Results revealed a number of rare sequence variants in genes related to elastic fibre assembly and integrity. For instance, the involvement of fibrillins and collagen type VI in the formation of a modified microfibrillar scaffold may lead to elastic fibres less resilient and more prone to hydroxyapatite deposition. Moreover, data reveal that changes in mitochondrial metabolic pathways are sustained by a genetic background and emphasize that a persistent chronic oxidative stress can further influence extracellular matrix homeostasis and cell signalling through the TGFß-BMP axis. Eventually, the presence of multiple rare sequence variants in the Solute Carrier Family 25 Member 5 (SLC25A5) gene is suggestive of the role of this gene as a key factor linking mitochondria metabolism, ADP/ATP ratio and oxidative stress thus affecting extracellular matrix homeostasis and activation of pro-osteogenic factors.


Subject(s)
Calcinosis/genetics , Computational Biology/methods , Elastic Tissue , Extracellular Matrix Proteins/genetics , Extracellular Matrix/genetics , beta-Thalassemia/genetics , Calcinosis/metabolism , Elastic Tissue/metabolism , Elastic Tissue/pathology , Extracellular Matrix/metabolism , Extracellular Matrix Proteins/metabolism , Female , Humans , Male , Exome Sequencing/methods , beta-Thalassemia/metabolism
13.
Blood ; 131(17): 1960-1973, 2018 04 26.
Article in English | MEDLINE | ID: mdl-29519807

ABSTRACT

Naturally occurring, large deletions in the ß-globin locus result in hereditary persistence of fetal hemoglobin, a condition that mitigates the clinical severity of sickle cell disease (SCD) and ß-thalassemia. We designed a clustered regularly interspaced short palindromic repeats (CRISPR) and CRISPR-associated protein 9 (Cas9) (CRISPR/Cas9) strategy to disrupt a 13.6-kb genomic region encompassing the δ- and ß-globin genes and a putative γ-δ intergenic fetal hemoglobin (HbF) silencer. Disruption of just the putative HbF silencer results in a mild increase in γ-globin expression, whereas deletion or inversion of a 13.6-kb region causes a robust reactivation of HbF synthesis in adult erythroblasts that is associated with epigenetic modifications and changes in chromatin contacts within the ß-globin locus. In primary SCD patient-derived hematopoietic stem/progenitor cells, targeting the 13.6-kb region results in a high proportion of γ-globin expression in erythroblasts, increased HbF synthesis, and amelioration of the sickling cell phenotype. Overall, this study provides clues for a potential CRISPR/Cas9 genome editing approach to the therapy of ß-hemoglobinopathies.


Subject(s)
Anemia, Sickle Cell , CRISPR-Cas Systems , Fetal Hemoglobin , Gene Editing , Genetic Loci , Hematopoietic Stem Cells/metabolism , beta-Globins/genetics , Anemia, Sickle Cell/genetics , Anemia, Sickle Cell/metabolism , Anemia, Sickle Cell/pathology , Anemia, Sickle Cell/therapy , Cell Line , Fetal Hemoglobin/biosynthesis , Fetal Hemoglobin/genetics , Hematopoietic Stem Cells/pathology , Humans , beta-Globins/metabolism
14.
Haematologica ; 103(5): 778-786, 2018 05.
Article in English | MEDLINE | ID: mdl-29472357

ABSTRACT

Sickle cell disease is characterized by chronic anemia and vaso-occlusive crises, which eventually lead to multi-organ damage and premature death. Hematopoietic stem cell transplantation is the only curative treatment but it is limited by toxicity and poor availability of HLA-compatible donors. A gene therapy approach based on the autologous transplantation of lentiviral-corrected hematopoietic stem and progenitor cells was shown to be efficacious in one patient. However, alterations of the bone marrow environment and properties of the red blood cells hamper the harvesting and immunoselection of patients' stem cells from bone marrow. The use of Filgrastim to mobilize large numbers of hematopoietic stem and progenitor cells into the circulation has been associated with severe adverse events in sickle cell patients. Thus, broader application of the gene therapy approach requires the development of alternative mobilization methods. We set up a phase I/II clinical trial whose primary objective was to assess the safety of a single injection of Plerixafor in sickle cell patients undergoing red blood cell exchange to decrease the hemoglobin S level to below 30%. The secondary objective was to measure the efficiency of mobilization and isolation of hematopoietic stem and progenitor cells. No adverse events were observed. Large numbers of CD34+ cells were mobilized extremely quickly. Importantly, the mobilized cells contained high numbers of hematopoietic stem cells, expressed high levels of stemness genes, and engrafted very efficiently in immunodeficient mice. Thus, Plerixafor can be safely used to mobilize hematopoietic stem cells in sickle cell patients; this finding opens up new avenues for treatment approaches based on gene addition and genome editing. Clinicaltrials.gov identifier: NCT02212535.


Subject(s)
Anemia, Sickle Cell/therapy , Blood Transfusion , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Heterocyclic Compounds/administration & dosage , Anemia, Sickle Cell/metabolism , Anemia, Sickle Cell/pathology , Anti-HIV Agents/administration & dosage , Antigens, CD34/metabolism , Antisickling Agents/administration & dosage , Benzylamines , Case-Control Studies , Cells, Cultured , Cohort Studies , Cyclams , Hematopoietic Stem Cells/cytology , Humans , Hydroxyurea/administration & dosage
15.
Nature ; 551(7680): 327-332, 2017 11 16.
Article in English | MEDLINE | ID: mdl-29144448

ABSTRACT

Junctional epidermolysis bullosa (JEB) is a severe and often lethal genetic disease caused by mutations in genes encoding the basement membrane component laminin-332. Surviving patients with JEB develop chronic wounds to the skin and mucosa, which impair their quality of life and lead to skin cancer. Here we show that autologous transgenic keratinocyte cultures regenerated an entire, fully functional epidermis on a seven-year-old child suffering from a devastating, life-threatening form of JEB. The proviral integration pattern was maintained in vivo and epidermal renewal did not cause any clonal selection. Clonal tracing showed that the human epidermis is sustained not by equipotent progenitors, but by a limited number of long-lived stem cells, detected as holoclones, that can extensively self-renew in vitro and in vivo and produce progenitors that replenish terminally differentiated keratinocytes. This study provides a blueprint that can be applied to other stem cell-mediated combined ex vivo cell and gene therapies.


Subject(s)
Epidermal Cells , Epidermolysis Bullosa, Junctional/therapy , Regeneration , Stem Cells/cytology , Stem Cells/metabolism , Transgenes/genetics , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Differentiation , Cell Lineage , Cell Self Renewal , Cell Tracking , Child , Clone Cells/cytology , Clone Cells/metabolism , Dermis/cytology , Dermis/pathology , Epidermis/pathology , Epidermolysis Bullosa, Junctional/genetics , Epidermolysis Bullosa, Junctional/metabolism , Epidermolysis Bullosa, Junctional/pathology , Humans , Keratinocytes/cytology , Keratinocytes/metabolism , Keratinocytes/transplantation , Male , Proviruses/genetics , Kalinin
16.
Stem Cells Transl Med ; 6(12): 2106-2114, 2017 12.
Article in English | MEDLINE | ID: mdl-29080249

ABSTRACT

Hematopoiesis is the process of blood cell formation starting from hematopoietic stem/progenitor cells (HSPCs). The understanding of regulatory networks involved in hematopoiesis and their impact on gene expression is crucial to decipher the molecular mechanisms that control hematopoietic development in physiological and pathological conditions, and to develop novel therapeutic strategies. An increasing number of epigenetic studies aim at defining, on a genome-wide scale, the cis-regulatory sequences (e.g., promoters and enhancers) used by human HSPCs and their lineage-restricted progeny at different stages of development. In parallel, human genetic studies allowed the discovery of genetic variants mapping to cis-regulatory elements and associated with hematological phenotypes and diseases. Here, we summarize recent epigenetic and genetic studies in hematopoietic cells that give insights into human hematopoiesis and provide a knowledge basis for the development of novel therapeutic approaches. As an example, we discuss the therapeutic approaches targeting cis-regulatory regions to reactivate fetal hemoglobin for the treatment of ß-hemoglobinopathies. Epigenetic studies allowed the definition of cis-regulatory sequences used by human hematopoietic cells. Promoters and enhancers are targeted by transcription factors and are characterized by specific histone modifications. Genetic variants mapping to cis-regulatory elements are often associated with hematological phenotypes and diseases. In some cases, these variants can alter the binding of transcription factors, thus changing the expression of the target genes. Targeting cis-regulatory sequences represents a promising therapeutic approach for many hematological diseases. Stem Cells Translational Medicine 2017;6:2106-2114.


Subject(s)
Epigenesis, Genetic , Hematopoiesis/genetics , Animals , Gene Expression Regulation, Developmental , Genetic Therapy/methods , Humans
17.
J Vis Exp ; (123)2017 05 28.
Article in English | MEDLINE | ID: mdl-28605390

ABSTRACT

Moloney murine leukemia (MLV) virus-based retroviral vectors integrate predominantly in acetylated enhancers and promoters. For this reason, mLV integration sites can be used as functional markers of active regulatory elements. Here, we present a retroviral scanning tool, which allows the genome-wide identification of cell-specific enhancers and promoters. Briefly, the target cell population is transduced with an mLV-derived vector and genomic DNA is digested with a frequently cutting restriction enzyme. After ligation of genomic fragments with a compatible DNA linker, linker-mediated polymerase chain reaction (LM-PCR) allows the amplification of the virus-host genome junctions. Massive sequencing of the amplicons is used to define the mLV integration profile genome-wide. Finally, clusters of recurrent integrations are defined to identify cell-specific regulatory regions, responsible for the activation of cell-type specific transcriptional programs. The retroviral scanning tool allows the genome-wide identification of cell-specific promoters and enhancers in prospectively isolated target cell populations. Notably, retroviral scanning represents an instrumental technique for the retrospective identification of rare populations (e.g. somatic stem cells) that lack robust markers for prospective isolation.


Subject(s)
Regulatory Sequences, Nucleic Acid , Retroviridae/genetics , Virus Integration , Humans
18.
Stem Cell Reports ; 6(4): 618-632, 2016 Apr 12.
Article in English | MEDLINE | ID: mdl-27050947

ABSTRACT

Human skin is maintained by the differentiation and maturation of interfollicular stem and progenitors cells. We used DeepCAGE, genome-wide profiling of histone modifications and retroviral integration analysis, to map transcripts, promoters, enhancers, and super-enhancers (SEs) in prospectively isolated keratinocytes and transit-amplifying progenitors, and retrospectively defined keratinocyte stem cells. We show that >95% of the active promoters are in common and differentially regulated in progenitors and differentiated keratinocytes, while approximately half of the enhancers and SEs are stage specific and account for most of the epigenetic changes occurring during differentiation. Transcription factor (TF) motif identification and correlation with TF binding site maps allowed the identification of TF circuitries acting on enhancers and SEs during differentiation. Overall, our study provides a broad, genome-wide description of chromatin dynamics and differential enhancer and promoter usage during epithelial differentiation, and describes a novel approach to identify active regulatory elements in rare stem cell populations.


Subject(s)
Cell Differentiation/genetics , Epigenesis, Genetic , Keratinocytes/metabolism , Stem Cells/metabolism , Transcription, Genetic , Animals , Binding Sites/genetics , Cells, Cultured , Enhancer Elements, Genetic/genetics , Epidermal Cells , Foreskin/cytology , Gene Expression Profiling/methods , Gene Ontology , Gene Regulatory Networks , Histones/metabolism , Humans , Keratinocytes/cytology , Male , Mice , Models, Genetic , NIH 3T3 Cells , Promoter Regions, Genetic/genetics , Protein Binding , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/cytology , Transcription Factors/metabolism
19.
Sci Rep ; 6: 24724, 2016 Apr 20.
Article in English | MEDLINE | ID: mdl-27095295

ABSTRACT

Genome-wide approaches allow investigating the molecular circuitry wiring the genetic and epigenetic programs of human somatic stem cells. Hematopoietic stem/progenitor cells (HSPC) give rise to the different blood cell types; however, the molecular basis of human hematopoietic lineage commitment is poorly characterized. Here, we define the transcriptional and epigenetic profile of human HSPC and early myeloid and erythroid progenitors by a combination of Cap Analysis of Gene Expression (CAGE), ChIP-seq and Moloney leukemia virus (MLV) integration site mapping. Most promoters and transcripts were shared by HSPC and committed progenitors, while enhancers and super-enhancers consistently changed upon differentiation, indicating that lineage commitment is essentially regulated by enhancer elements. A significant fraction of CAGE promoters differentially expressed upon commitment were novel, harbored a chromatin enhancer signature, and may identify promoters and transcribed enhancers driving cell commitment. MLV-targeted genomic regions co-mapped with cell-specific active enhancers and super-enhancers. Expression analyses, together with an enhancer functional assay, indicate that MLV integration can be used to identify bona fide developmentally regulated enhancers. Overall, this study provides an overview of transcriptional and epigenetic changes associated to HSPC lineage commitment, and a novel signature for regulatory elements involved in cell identity.


Subject(s)
Cell Differentiation/genetics , Cell Lineage/genetics , Epigenesis, Genetic , Gene Expression Regulation, Viral , Hematopoietic Stem Cells/cytology , Regulatory Sequences, Nucleic Acid , Retroviridae/genetics , Transcriptome , Base Sequence , Consensus Sequence , Enhancer Elements, Genetic , Hematopoietic Stem Cells/metabolism , Histones/metabolism , Humans , Multipotent Stem Cells/cytology , Organ Specificity , Position-Specific Scoring Matrices , Promoter Regions, Genetic , Transcription Initiation, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...