Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters











Publication year range
1.
Mol Ther Methods Clin Dev ; 32(3): 101311, 2024 Sep 12.
Article in English | MEDLINE | ID: mdl-39234443

ABSTRACT

Lamellar ichthyosis (LI) is a chronic disease, mostly caused by mutations in the TGM1 gene, marked by impaired skin barrier formation. No definitive therapies are available, and current treatments aim at symptomatic relief. LI mouse models often fail to faithfully replicate the clinical and histopathological features of human skin conditions. To develop advanced therapeutic approaches, such as combined ex vivo cell and gene therapy, we established a human cellular model of LI by efficient CRISPR-Cas9-mediated gene ablation of the TGM1 gene in human primary clonogenic keratinocytes. Gene-edited cells showed complete absence of transglutaminase 1 (TG1) expression and recapitulated a hyperkeratotic phenotype with most of the molecular hallmarks of LI in vitro. Using a self-inactivating γ-retroviral (SINγ-RV) vector expressing transgenic TGM1 under the control of its own promoter, we tested an ex vivo gene therapy approach and validate the model of LI as a platform for pre-clinical evaluation studies. Gene-corrected TGM1-null keratinocytes displayed proper TG1 expression, enzymatic activity, and cornified envelope formation and, hence, restored proper epidermal architecture. Single-cell multiomics analysis demonstrated proviral integrations in holoclone-forming epidermal stem cells, which are crucial for epidermal regeneration. This study serves as a proof of concept for assessing the potential of this therapeutic approach in treating TGM1-dependent LI.

2.
Mol Ther ; 2024 Jul 22.
Article in English | MEDLINE | ID: mdl-39044427

ABSTRACT

Fetal hemoglobin (HbF) reactivation expression through CRISPR-Cas9 is a promising strategy for the treatment of sickle cell disease (SCD). Here, we describe a genome editing strategy leading to reactivation of HbF expression by targeting the binding sites (BSs) for the lymphoma-related factor (LRF) repressor in the γ-globin promoters. CRISPR-Cas9 treatment in healthy donor (HD) and patient-derived HSPCs resulted in a high frequency of LRF BS disruption and potent HbF synthesis in their erythroid progeny. LRF BS disruption did not impair HSPC engraftment and differentiation but was more efficient in SCD than in HD cells. However, SCD HSPCs showed a reduced engraftment and a myeloid bias compared with HD cells. We detected off-target activity and chromosomal rearrangements, particularly in SCD samples (likely because of the higher overall editing efficiency) but did not impact the target gene expression and HSPC engraftment and differentiation. Transcriptomic analyses showed that the editing procedure results in the up-regulation of genes involved in DNA damage and inflammatory responses, which was more evident in SCD HSPCs. This study provides evidence of efficacy and safety for an editing strategy based on HbF reactivation and highlights the need of performing safety studies in clinically relevant conditions, i.e., in patient-derived HSPCs.

3.
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
4.
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.

5.
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
6.
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
7.
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
8.
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
9.
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
10.
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.

SELECTION OF CITATIONS
SEARCH DETAIL