Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
J Vis Exp ; (197)2023 07 21.
Article in English | MEDLINE | ID: mdl-37590510

ABSTRACT

The lymphatic system participates in the regulation of immune surveillance, lipid absorption, and tissue fluid balance. The isolation of murine lymphatic endothelial cells is an important process for lymphatic research, as it allows the performance of in vitro and biochemical experiments on the isolated cells. Moreover, the development of Cre-lox technology has enabled the tissue-specific deficiency of genes that cannot be globally targeted, leading to the precise determination of their role in the studied tissues. The dissection of the role of certain genes in lymphatic physiology and pathophysiology requires the use of lymphatic-specific promoters, and thus, the experimental verification of the expression levels of the targeted genes. Methods for efficient isolation of lymphatic endothelial cells from wild-type or transgenic mice enable the use of ex vivo and in vitro assays to study the mechanisms regulating the lymphatic functions and the identification of the expression levels of the studied proteins. We have developed, standardized and present a protocol for the efficient isolation of murine dermal lymphatic endothelial cells (DLECs) via magnetic bead purification based on LYVE-1 expression. The protocol outlined aims to equip researchers with a tool to further understand and elucidate important players of lymphatic endothelial cell functions, especially in facilities where fluorescence-activated cell sorting equipment is not available.


Subject(s)
Endothelial Cells , Lymphatic Vessels , Mice , Animals , Mice, Transgenic , Dissection , Extracellular Fluid
2.
Viruses ; 14(12)2022 12 05.
Article in English | MEDLINE | ID: mdl-36560719

ABSTRACT

We have previously demonstrated that both the original γ-globin lentiviral vector (LV) GGHI and the optimized GGHI-mB-3D LV, carrying the novel regulatory elements of the 3D HPFH-1 enhancer and the 3' ß-globin UTR, can significantly increase HbF production in thalassemic CD34+ cells and ameliorate the disease phenotype in vitro. In the present study, we investigated whether the GGHI-mB-3D vector can also exhibit an equally therapeutic effect, following the transduction of sickle cell disease (SCD) CD34+ cells at MOI 100, leading to HbF increase coupled with HbS decrease, and thus, to phenotype improvement in vitro. We show that GGHI-mB-3D LV can lead to high and potentially therapeutic HbF levels, reaching a mean 2-fold increase to a mean value of VCN/cell of 1.0 and a mean transduction efficiency of 55%. Furthermore, this increase was accompanied by a significant 1.6-fold HbS decrease, a beneficial therapeutic feature for SCD. In summary, our data demonstrate the efficacy of the optimized γ-globin lentiviral vector to improve the SCD phenotype in vitro, and highlights its potential use in future clinical SCD trials.


Subject(s)
Anemia, Sickle Cell , beta-Thalassemia , Humans , gamma-Globins/genetics , Genetic Therapy , Fetal Hemoglobin/genetics , Genetic Vectors/genetics , Lentivirus/genetics , beta-Thalassemia/genetics , beta-Thalassemia/therapy , Anemia, Sickle Cell/genetics , Anemia, Sickle Cell/therapy
3.
Cell Rep ; 31(9): 107688, 2020 06 02.
Article in English | MEDLINE | ID: mdl-32492433

ABSTRACT

Leukemia stem cells (LSCs) are believed to have more distinct vulnerabilities than the bulk acute myeloid leukemia (AML) cells, but their rarity and the lack of universal markers for their prospective isolation hamper their study. We report that genetically clonal induced pluripotent stem cells (iPSCs) derived from an AML patient and characterized by exceptionally high engraftment potential give rise, upon hematopoietic differentiation, to a phenotypic hierarchy. Through fate-tracking experiments, xenotransplantation, and single-cell transcriptomics, we identify a cell fraction (iLSC) that can be isolated prospectively by means of adherent in vitro growth that resides on the apex of this hierarchy and fulfills the hallmark features of LSCs. Through integrative genomic studies of the iLSC transcriptome and chromatin landscape, we derive an LSC gene signature that predicts patient survival and uncovers a dependency of LSCs, across AML genotypes, on the RUNX1 transcription factor. These findings can empower efforts to therapeutically target AML LSCs.


Subject(s)
Core Binding Factor Alpha 2 Subunit/metabolism , Leukemia, Myeloid, Acute/pathology , Animals , Cell Differentiation , Cell Line , Chromatin/metabolism , Core Binding Factor Alpha 2 Subunit/antagonists & inhibitors , Core Binding Factor Alpha 2 Subunit/genetics , Gene Expression Regulation , Genetic Heterogeneity , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Leukemia, Myeloid, Acute/metabolism , Markov Chains , Mice , Mice, Inbred NOD , Mice, SCID , Phenotype , RNA Interference , RNA, Small Interfering/metabolism , RNA-Seq , Single-Cell Analysis
4.
Dis Model Mech ; 12(6)2019 06 04.
Article in English | MEDLINE | ID: mdl-31171568

ABSTRACT

Induced pluripotent stem cells (iPSCs) are derived from somatic cells through a reprogramming process, which converts them to a pluripotent state, akin to that of embryonic stem cells. Over the past decade, iPSC models have found increasing applications in the study of human diseases, with blood disorders featuring prominently. Here, we discuss methodological aspects pertaining to iPSC generation, hematopoietic differentiation and gene editing, and provide an overview of uses of iPSCs in modeling the cell and gene therapy of inherited genetic blood disorders, as well as their more recent use as models of myeloid malignancies. We also discuss the strengths and limitations of iPSCs compared to model organisms and other cellular systems commonly used in hematology research.


Subject(s)
Hematologic Diseases/pathology , Induced Pluripotent Stem Cells/cytology , Models, Biological , Animals , Disease Models, Animal , Genetic Engineering , Hematopoiesis , Humans
5.
Hum Gene Ther ; 30(5): 601-617, 2019 05.
Article in English | MEDLINE | ID: mdl-30324804

ABSTRACT

It has previously been demonstrated that the self-inactivating γ-globin lentiviral vector GGHI can significantly increase fetal hemoglobin (HbF) in erythroid cells from thalassemia patients and thus improve the disease phenotype in vitro. In the present study, the GGHI vector was improved further by incorporating novel enhancer elements and also pseudotyping it with the baboon endogenous virus envelope glycoprotein BaEVRless, which efficiently and specifically targets human CD34+ cells. We evaluated the hypothesis that the newly constructed vector designated as GGHI-mB-3D would increase hCD34+ cell tropism and thus transduction efficiency at low multiplicity of infection, leading to increased transgene expression. High and stable HbF expression was demonstrated in thalassemic cells for the resulting GGHI-mB-3D/BaEVRless vector, exhibiting increased transduction efficiency compared to the original GGHI-mB-3D/VSVG vector, with a concomitant 91% mean HbF increase at a mean vector copy number per cell of 0.86 and a mean transduction efficiency of 56.4%. Transduced populations also exhibited a trend toward late erythroid, orthochromatic differentiation and reduced apoptosis, a further indication of successful gene therapy treatment. Monitoring expression of ATG5, a key link between autophagy and apoptosis, it was established that this correction correlates with a reduction of enhanced autophagy activation, a typical feature of thalassemic polychromatophilic normoblasts. This work provides novel mechanistic insights into gene therapy-mediated correction of erythropoiesis and demonstrates the beneficial role of BaEVRless envelope glycoprotein compared to VSVG pseudotyping and of the novel GGHI-mB-3D/BaEVRless lentiviral vector for enhanced thalassemia gene therapy.


Subject(s)
Erythropoiesis/genetics , Gene Expression , Genetic Vectors/genetics , Lentivirus/genetics , Transgenes , beta-Thalassemia/genetics , gamma-Globins/genetics , Fetal Hemoglobin/genetics , Gene Order , Gene Transfer Techniques , Genetic Engineering , Genetic Therapy/methods , Hematopoietic Stem Cells/metabolism , Humans , Recombination, Genetic , Transduction, Genetic , beta-Thalassemia/therapy
6.
Stem Cell Reports ; 10(5): 1610-1624, 2018 05 08.
Article in English | MEDLINE | ID: mdl-29681544

ABSTRACT

Connecting specific cancer genotypes with phenotypes and drug responses constitutes the central premise of precision oncology but is hindered by the genetic complexity and heterogeneity of primary cancer cells. Here, we use patient-derived induced pluripotent stem cells (iPSCs) and CRISPR/Cas9 genome editing to dissect the individual contributions of two recurrent genetic lesions, the splicing factor SRSF2 P95L mutation and the chromosome 7q deletion, to the development of myeloid malignancy. Using a comprehensive panel of isogenic iPSCs-with none, one, or both genetic lesions-we characterize their relative phenotypic contributions and identify drug sensitivities specific to each one through a candidate drug approach and an unbiased large-scale small-molecule screen. To facilitate drug testing and discovery, we also derive SRSF2-mutant and isogenic normal expandable hematopoietic progenitor cells. We thus describe here an approach to dissect the individual effects of two cooperating mutations to clinically relevant features of malignant diseases.


Subject(s)
Antineoplastic Agents/therapeutic use , Induced Pluripotent Stem Cells/pathology , Mutation/genetics , Neoplasms/drug therapy , Neoplasms/genetics , Alternative Splicing/genetics , Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Humans , Induced Pluripotent Stem Cells/drug effects , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/pathology , Neoplasms/pathology , Phenotype , Serine-Arginine Splicing Factors/genetics , Serine-Arginine Splicing Factors/metabolism , Small Molecule Libraries/pharmacology
7.
Virus Res ; 175(1): 1-11, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23583684

ABSTRACT

Gene therapy utilizing lentiviral vectors (LVs) constitutes a real therapeutic alternative for many inherited monogenic diseases. Therefore, the generation of functional vectors using fast, non-laborious and cost-effective strategies is imperative. Among the available concentration methods for VSV-G pseudotyped lentiviruses to achieve high therapeutic titers, ultracentrifugation represents the most common approach. However, the procedure requires special handling and access to special instrumentation, it is time-consuming, and most importantly, it is cost-ineffective due to the high maintenance expenses and consumables of the ultracentrifuge apparatus. Here we describe an improved protocol in which vector stocks are prepared by transient transfection using standard cell culture media and are then concentrated by ultrafiltration, resulting in functional vector titers of up to 6×10(9) transducing units per millilitre (TU/ml) without the involvement of any purification step. Although ultrafiltration per se for concentrating viruses is not a new procedure, our work displays one major novelty; we characterized the nature and the constituents of the viral batches produced by ultrafiltration using peptide mass fingerprint analysis. We also determined the viral functional titer by employing flow cytometry and evaluated the actual viral particle size and concentration in real time by using laser-based nanoparticle tracking analysis based on Brownian motion. Vectors generated by this production method are contained in intact virions and when tested to transduce in vitro either murine total bone marrow or human CD34(+) hematopoietic stem cells, resulted in equal transduction efficiency and reduced toxicity, compared to lentiviral vectors produced using standard ultracentrifugation-based methods. The data from this study can eventually lead to the improvement of protocols and technical modifications for the clinical trials for gene therapy.


Subject(s)
Genetic Vectors/isolation & purification , Lentivirus/isolation & purification , Ultrafiltration/methods , Virology/methods , Animals , Genetic Therapy/methods , Genetic Vectors/chemistry , Hematopoietic Stem Cells/virology , Humans , Lentivirus/chemistry , Mice , Mice, Inbred C57BL , Peptide Mapping , Transduction, Genetic , Ultracentrifugation/methods
8.
Hum Gene Ther ; 23(1): 15-31, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21875313

ABSTRACT

To address how low titer, variable expression, and gene silencing affect gene therapy vectors for hemoglobinopathies, in a previous study we successfully used the HPFH (hereditary persistence of fetal hemoglobin)-2 enhancer in a series of oncoretroviral vectors. On the basis of these data, we generated a novel insulated self-inactivating (SIN) lentiviral vector, termed GGHI, carrying the (A)γ-globin gene with the -117 HPFH point mutation and the HPFH-2 enhancer and exhibiting a pancellular pattern of (A)γ-globin gene expression in MEL-585 clones. To assess the eventual clinical feasibility of this vector, GGHI was tested on CD34(+) hematopoietic stem cells from nonmobilized peripheral blood or bone marrow from 20 patients with ß-thalassemia. Our results show that GGHI increased the production of γ-globin by 32.9% as measured by high-performance liquid chromatography (p=0.001), with a mean vector copy number per cell of 1.1 and a mean transduction efficiency of 40.3%. Transduced populations also exhibited a lower rate of apoptosis and resulted in improvement of erythropoiesis with a higher percentage of orthochromatic erythroblasts. This is the first report of a locus control region (LCR)-free SIN insulated lentiviral vector that can be used to efficiently produce the anticipated therapeutic levels of γ-globin protein in the erythroid progeny of primary human thalassemic hematopoietic stem cells in vitro.


Subject(s)
Fetal Hemoglobin/metabolism , Genetic Therapy/methods , Hematopoietic Stem Cells/metabolism , Lentivirus/metabolism , Virus Inactivation , beta-Thalassemia/therapy , Antigens, CD34/metabolism , Apoptosis , Chromatography, High Pressure Liquid , Cloning, Molecular , Enhancer Elements, Genetic , Erythroid Cells/metabolism , Erythropoiesis , Fetal Hemoglobin/genetics , Gene Transfer Techniques , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Genetic Vectors/metabolism , HEK293 Cells , Hematopoietic Stem Cells/pathology , Humans , Lentivirus/genetics , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Plasmids/genetics , Plasmids/metabolism , Point Mutation , Promoter Regions, Genetic , Transfection , Vesicular stomatitis Indiana virus/genetics , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , gamma-Globins/genetics , gamma-Globins/metabolism
9.
Blood ; 117(10): 2817-26, 2011 Mar 10.
Article in English | MEDLINE | ID: mdl-21156846

ABSTRACT

ß-Thalassemia major results from severely reduced or absent expression of the ß-chain of adult hemoglobin (α2ß2;HbA). Increased levels of fetal hemoglobin (α2γ2;HbF), such as occurs with hereditary persistence of HbF, ameliorate the severity of ß-thalassemia, raising the potential for genetic therapy directed at enhancing HbF. We used an in vitro model of human erythropoiesis to assay for enhanced production of HbF after gene delivery into CD34(+) cells obtained from mobilized peripheral blood of normal adults or steady-state bone marrow from patients with ß-thalassemia major. Lentiviral vectors encoding (1) a human γ-globin gene with or without an insulator, (2) a synthetic zinc-finger transcription factor designed to interact with the γ-globin gene promoters, or (3) a short-hairpin RNA targeting the γ-globin gene repressor, BCL11A, were tested. Erythroid progeny of normal CD34(+) cells demonstrated levels of HbF up to 21% per vector copy. For ß-thalassemic CD34(+) cells, similar gene transfer efficiencies achieved HbF production ranging from 45% to 60%, resulting in up to a 3-fold increase in the total cellular Hb content. These observations suggest that both lentiviral-mediated γ-globin gene addition and genetic reactivation of endogenous γ-globin genes have potential to provide therapeutic HbF levels to patients with ß-globin deficiency.


Subject(s)
Erythroid Precursor Cells/metabolism , Fetal Hemoglobin/biosynthesis , Gene Transfer Techniques , Genetic Therapy , beta-Thalassemia/therapy , gamma-Globins/genetics , Antigens, CD34/metabolism , Blotting, Southern , Blotting, Western , Cell Separation , Erythropoiesis/physiology , Fetal Hemoglobin/genetics , Flow Cytometry , Genetic Vectors , Humans , Lentivirus/genetics , Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...