Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
bioRxiv ; 2024 Mar 14.
Article in English | MEDLINE | ID: mdl-38559013

ABSTRACT

Metachromatic leukodystrophy (MLD) is a fatal lysosomal storage disease (LSD) characterized by the deficient enzymatic activity of arylsulfatase A (ARSA). Combined autologous hematopoietic stem cell transplant (HSCT) with lentiviral (LV) based gene therapy has great potential to treat MLD. However, if enzyme production is inadequate, this could result in continued loss of motor function, implying a high vector copy number (VCN) requirement for optimal enzymatic output. This may place children at increased risk for genomic toxicity due to higher VCN. We increased the expression of ARSA cDNA at single integration by generating novel LVs, optimizing ARSA expression, and enhancing safety. In addition, our vectors achieved optimal transduction in mouse and human HSC with minimal multiplicity of infection (MOI). Our top-performing vector (EA1) showed at least 4X more ARSA activity than the currently EU-approved vector and a superior ability to secrete vesicle-associated ARSA, a critical modality to transfer functional enzymes from microglia to oligodendrocytes. Three-month-old Arsa -KO MLD mice transplanted with Arsa -KO BM cells transduced with 0.6 VCN of EA1 demonstrated behavior and CNS histology matching WT mice. Our novel vector boosts efficacy while improving safety as a robust approach for treating early symptomatic MLD patients.

3.
Blood Adv ; 7(22): 6873-6885, 2023 11 28.
Article in English | MEDLINE | ID: mdl-37672319

ABSTRACT

ß-thalassemias are common hemoglobinopathies due to mutations in the ß-globin gene that lead to hemolytic anemias. Premature death of ß-thalassemic erythroid precursors results in ineffective erythroid maturation, increased production of erythropoietin (EPO), expansion of erythroid progenitor compartment, extramedullary erythropoiesis, and splenomegaly. However, the molecular mechanism of erythroid apoptosis in ß-thalassemia is not well understood. Using a mouse model of ß-thalassemia (Hbbth3/+), we show that dysregulated expression of the FOXO3 transcription factor is implicated in ß-thalassemia erythroid apoptosis. In Foxo3-/-/Hbbth3/+ mice, erythroid apoptosis is significantly reduced, whereas erythroid cell maturation, and red blood cell and hemoglobin production are substantially improved even with elevated reactive oxygen species in double-mutant erythroblasts. However, persistence of elevated reticulocytes and splenomegaly suggests that ineffective erythropoiesis is not resolved in Foxo3-/-/Hbbth3/+. We found the cell cycle inhibitor Cdkn1a (cyclin-dependent kinase inhibitor p21), a FOXO3 target gene, is markedly upregulated in both mouse and patient-derived ß-thalassemic erythroid precursors. Double-mutant p21/Hbbth3/+ mice exhibited embryonic lethality with only a fraction of mice surviving to weaning. Notably, studies in adult mice displayed greatly reduced apoptosis and circulating Epo in erythroid compartments of surviving p21-/-/Hbbth3/+ mice relative to Hbbth3/+ mice, whereas ineffective erythroid cell maturation, extramedullary erythropoiesis, and splenomegaly were not modified. These combined results suggest that mechanisms that control ß-thalassemic erythroid cell survival and differentiation are uncoupled from ineffective erythropoiesis and involve a molecular network including FOXO3 and P21. Overall, these studies provide a new framework for investigating ineffective erythropoiesis in ß-thalassemia.


Subject(s)
Erythropoiesis , beta-Thalassemia , Humans , Apoptosis , beta-Thalassemia/genetics , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Erythropoiesis/genetics , Splenomegaly
4.
Science ; 381(6656): 436-443, 2023 07 28.
Article in English | MEDLINE | ID: mdl-37499029

ABSTRACT

Hematopoietic stem cells (HSCs) are the source of all blood cells over an individual's lifetime. Diseased HSCs can be replaced with gene-engineered or healthy HSCs through HSC transplantation (HSCT). However, current protocols carry major side effects and have limited access. We developed CD117/LNP-messenger RNA (mRNA), a lipid nanoparticle (LNP) that encapsulates mRNA and is targeted to the stem cell factor receptor (CD117) on HSCs. Delivery of the anti-human CD117/LNP-based editing system yielded near-complete correction of hematopoietic sickle cells. Furthermore, in vivo delivery of pro-apoptotic PUMA (p53 up-regulated modulator of apoptosis) mRNA with CD117/LNP affected HSC function and permitted nongenotoxic conditioning for HSCT. The ability to target HSCs in vivo offers a nongenotoxic conditioning regimen for HSCT, and this platform could be the basis of in vivo genome editing to cure genetic disorders, which would abrogate the need for HSCT.


Subject(s)
Gene Editing , Hematopoietic Stem Cells , Proto-Oncogene Proteins c-kit , RNA, Messenger , Gene Editing/methods , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/metabolism , Proto-Oncogene Proteins c-kit/genetics , RNA, Messenger/genetics , Animals , Humans , Mice
6.
Int J Paleopathol ; 33: 25-29, 2021 06.
Article in English | MEDLINE | ID: mdl-33640560

ABSTRACT

OBJECTIVE: This study presents evidence of a probable case of holoprosencephaly with cyclopia, which has been rarely reported in the paleopathological literature. MATERIALS: The skeletal remains of a male fetus between 36 and 40 gestational weeks from the Collezione Antropologica LABANOF (CAL) Milano Cemetery Skeletal Collection were studied. METHODS: The bones were macroscopically examined, and pathological anomalies were recorded and evaluated alongside paleopathological and clinical literature. RESULTS: Developmental anomalies were observed. In particular, a single orbit and optical canal were present, and the frontal, sphenoid and palatine bones were prematurely fused. These changes altered the normal morphology of the midline structures of the cranium and face. CONCLUSIONS: The developmental anomalies observed are consistent with a case of holoprosencephaly associated with cyclopia. SIGNIFICANCE: Holoprosencephaly is a fatal congenital condition caused by the failure of the prosencephalon to separate in two halves. This condition is clinically well-known, with an estimated modern incidence of 1/16,000 births; however, the paleopathological literature lacks reports that would help anthropologists and paleopathologists interpret these anomalous signs on dry bone. This report documents a rare paleopathological case of the condition on a full-term fetus from a modern skeletal collection. LIMITATIONS: Taphonomic and anthropic factors may have impaired the observation of all pathological features. SUGGESTIONS FOR FURTHER RESEARCH: Comparative studies with cases from documented collections could improve knowledge of the appearance of this condition on dry bones.


Subject(s)
Holoprosencephaly , Cemeteries , Fetus , Humans , Male , Skull/diagnostic imaging
7.
Mol Ther ; 29(4): 1625-1638, 2021 04 07.
Article in English | MEDLINE | ID: mdl-33515514

ABSTRACT

Ongoing clinical trials for treatment of beta-globinopathies by gene therapy involve the transfer of the beta-globin gene, which requires integration of three to four copies per genome in most target cells. This high proviral load may increase genome toxicity, potentially limiting the safety of this therapy and relegating its use to total body myeloablation. We hypothesized that introducing an additional hypersensitive site from the locus control region, the complete sequence of the second intron of the beta-globin gene, and the ankyrin insulator may enhance beta-globin expression. We identified a construct, ALS20, that synthesized significantly higher adult hemoglobin levels than those of other constructs currently used in clinical trials. These findings were confirmed in erythroblastic cell lines and in primary cells isolated from sickle cell disease patients. Bone marrow transplantation studies in beta-thalassemia mice revealed that ALS20 was curative at less than one copy per genome. Injection of human CD34+ cells transduced with ALS20 led to safe, long-term, and high polyclonal engraftment in xenograft experiments. Successful treatment of beta-globinopathies with ALS20 could potentially be achieved at less than two copies per genome, minimizing the risk of cytotoxic events and lowering the intensity of myeloablation.


Subject(s)
Anemia, Sickle Cell/genetics , Bone Marrow Transplantation , Genetic Therapy , beta-Globins/genetics , beta-Thalassemia/genetics , Anemia, Sickle Cell/blood , Anemia, Sickle Cell/pathology , Anemia, Sickle Cell/therapy , Animals , Gene Expression/genetics , Genetic Vectors/genetics , Genetic Vectors/pharmacology , Hemoglobins/genetics , Heterografts , Humans , Lentivirus/genetics , Locus Control Region/genetics , Mice , Transduction, Genetic , beta-Globins/therapeutic use , beta-Thalassemia/blood , beta-Thalassemia/pathology , beta-Thalassemia/therapy
8.
Leg Med (Tokyo) ; 48: 101801, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33268276

ABSTRACT

In forensic anthropology, correct identification of human deciduous teeth is of paramount importance for age-at-death estimation and relies on detailed anatomical descriptions. Yet literature is scarce on indications: details on the morphology of molar tooth germs of fetuses and newborns, developing from multiple mineralized centers that will eventually coalesce, are scant. This paper presents new anatomical elements for practitioners to identify human molar tooth germs at early developmental stages. 126 deciduous molars from 22 modern skeletons of fetuses and newborns (with a known age-at-death ranging between 0 days and 2 months and 21 days postnatal), without reported or observed dental pathological signs, were selected from the Collezione Antropologica LABANOF (CAL) documented skeletal collection. Gross anatomical descriptions of the morphology and configuration of the centers were provided, considering the number of mineralized centers, the shape and the outline of the occlusal plane at different stages. Three different developmental stages were observed in the maxillary first and second molar and the mandibular first molar, whereas in the mandibular second molar four stages were observed. For each stage, we provide additional detailed morphological descriptions, sketches outlining the shape of the tooth germ, and a picture of the tooth; also, indications for siding the teeth are presented. This information can be used by forensic anthropologists and odontologists for a proper identification when tooth germs are not found in anatomical connection within the dental sockets. Further analyses that encompass more age groups on a larger sample would allow to map the entire crown development of deciduous molars.


Subject(s)
Age Determination by Teeth/methods , Forensic Anthropology/methods , Molar/anatomy & histology , Tooth Germ/anatomy & histology , Tooth, Deciduous/anatomy & histology , Humans , Infant, Newborn , Mandible , Maxilla , Molar/embryology , Tooth Crown/anatomy & histology , Tooth Crown/embryology , Tooth Crown/growth & development , Tooth Germ/embryology , Tooth, Deciduous/embryology
9.
Mediterr J Hematol Infect Dis ; 11(1): e2019067, 2019.
Article in English | MEDLINE | ID: mdl-31700592

ABSTRACT

Inherited hemoglobin disorders, including beta-thalassemia (BT) and sickle-cell disease (SCD), are the most common monogenic diseases worldwide, with a global carrier frequency of over 5%.1 With migration, they are becoming more common worldwide, making their management and care an increasing concern for health care systems. BT is characterized by an imbalance in the α/ß-globin chain ratio, ineffective erythropoiesis, chronic hemolytic anemia, and compensatory hemopoietic expansion.1 Globally, there are over 25,000 births each year with transfusion-dependent thalassemia (TDT). The currently available treatment for TDT is lifelong transfusions and iron chelation therapy or allogenic bone marrow transplantation as a curative option. SCD affects 300 million people worldwide2 and severely impacts the quality of life of patients who experience unpredictable, recurrent acute and chronic severe pain, stroke, infections, pulmonary disease, kidney disease, retinopathy, and other complications. While survival has been dramatically extended, quality of life is markedly reduced by disease- and treatment-associated morbidity. The development of safe, tissue-specific and efficient vectors, and efficient gene-editing technologies have led to the development of several gene therapy trials for BT and SCD. However, the complexity of the approach presents its hurdles. Fundamental factors at play include the requirement for myeloablation on a patient with benign disease, the age of the patient, and the consequent bone marrow microenvironment. A successful path from proof-ofconcept studies to commercialization must render gene therapy a sustainable and accessible approach for a large number of patients. Furthermore, the cost of these therapies is a considerable challenge for the health care system. While new promising therapeutic options are emerging,3,4 and many others are on the pipeline,5 gene therapy can potentially cure patients. We herein provide an overview of the most recent, likely potentially curative therapies for hemoglobinopathies and a summary of the challenges that these approaches entail.

10.
Anal Bioanal Chem ; 411(29): 7669-7680, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31273412

ABSTRACT

There is a general agreement that pharmacologically mediated stimulation of human γ-globin gene expression and increase of production of fetal hemoglobin (HbF) is a potential therapeutic approach in the experimental therapy of ß-thalassemia and sickle cell anemia. Here, we report the development and characterization of cellular biosensors carrying enhanced green fluorescence protein (EGFP) and red fluorescence protein (RFP) genes under the control of the human γ-globin and ß-globin gene promoters, respectively; these dual-reporter cell lines are suitable to identify the induction ability of screened compounds on the transcription in erythroid cells of γ-globin and ß-globin genes by FACS with efficiency and reproducibility. Our experimental system allows to identify (a) HbF inducers stimulating to different extent the activity of the γ-globin gene promoter and (b) molecules that stimulate also the activity of the ß-globin gene promoter. A good correlation does exist between the results obtained by using the EGFP/RFP clones and experiments performed on erythroid precursor cells from ß-thalassemic patients, confirming that this experimental system can be employed for high-throughput screening (HTS) analysis. Finally, we have demonstrated that this dual-reporter cell line can be used for HTS in 384-well plate, in order to identify novel HbF inducers for the therapy of ß-thalassemia and sickle cell anemia. Graphical abstract.


Subject(s)
Biosensing Techniques , Cell Differentiation/drug effects , Erythrocytes/drug effects , High-Throughput Screening Assays/methods , Promoter Regions, Genetic , Transcription, Genetic , beta-Globins/genetics , gamma-Globins/genetics , Erythrocytes/cytology , Fetal Hemoglobin/genetics , Green Fluorescent Proteins/genetics , Humans , K562 Cells , Reproducibility of Results
12.
Mol Diagn Ther ; 23(2): 173-186, 2019 04.
Article in English | MEDLINE | ID: mdl-30701409

ABSTRACT

Inherited monogenic disorders such as beta-hemoglobinopathies (BH) are fitting candidates for treatment via gene therapy by gene transfer or gene editing. The reported safety and efficacy of lentiviral vectors in preclinical studies have led to the development of several clinical trials for the addition of a functional beta-globin gene. Across trials, dozens of transfusion-dependent patients with sickle cell disease (SCD) and transfusion-dependent beta-thalassemia (TDT) have been treated via gene therapy and have achieved reduced transfusion requirements. While overall results are encouraging, the outcomes appear to be strongly influenced by the level of lentiviral integration in transduced cells after engraftment, as well as the underlying genotype resulting in thalassemia. In addition, the method of procurement of hematopoietic stem cells can affect their quality and thus the outcome of gene therapy both in SCD and TDT. This suggests that new studies aimed at maximizing the number of corrected cells with long-term self-renewal potential are crucial to ensure successful treatment for every patient. Recent advancements in gene transfer and bone marrow transplantation have improved the success of this approach, and the results obtained by using these strategies demonstrated significant improvement of gene transfer outcome in patients. The advent of new gene-editing technologies has suggested additional therapeutic options. These are primarily focused on correcting the defective beta-globin gene or editing the expression of genes or genomic segments that regulate fetal hemoglobin synthesis. In this review, we aim to establish the potential benefits of gene therapy for BH, to summarize the status of the ongoing trials, and to discuss the possible improvement or direction for future treatments.


Subject(s)
Genetic Therapy , Hemoglobinopathies/genetics , Hemoglobinopathies/therapy , beta-Globins/genetics , Epigenesis, Genetic , Gene Editing , Humans , Treatment Outcome
13.
Haematologica ; 106(5): 1433-1442, 2019 May 01.
Article in English | MEDLINE | ID: mdl-32439726

ABSTRACT

ß-thalassemia is a disorder caused by altered hemoglobin protein synthesis and affects individuals worldwide. Severe forms of the disease, left untreated, can result in death before the age of 3 years (1). The standard of care consists of chronic and costly palliative treatment by blood transfusion combined with iron chelation. This dual approach suppresses anemia and reduces iron-related toxicities in patients. Allogeneic bone marrow transplant is an option, but limited by the availability of a highly compatible HSC donor. While gene therapy is been explored in several trials, its use is highly limited to developed regions with centers of excellence and well-established healthcare systems (2). Hence, there remains a tremendous unmet medical need to develop alternative treatment strategies for ß-thalassemia (3). Occurrence of aberrant splicing is one of the processes that affects ß-globin synthesis in ß-thalassemia. The (C>G) IVS-2-745 is a splicing mutation within intron 2 of the ß-globin gene. It leads to an aberrantly spliced mRNA that incorporates an intron fragment. This results in an in-frame premature termination codon that inhibits ß-globin production. Here, we propose the use of uniform 2'-O-methoxyethyl (2'-MOE) splice switching oligos (SSOs) to reverse this aberrant splicing in the pre-mRNA. With these lead SSOs we show aberrant to wild type splice switching. This switching leads to an increase of adult hemoglobin (HbA) up to 80% in erythroid cells from patients with the IVS-2-745 mutation. Furthermore, we demonstrate a restoration of the balance between ß-like- and α-globin chains, and up to an 87% reduction in toxic α-heme aggregates. While examining the potential benefit of 2'-MOE-SSOs in a mixed sickle-thalassemic phenotypic setting, we found reduced HbS synthesis and sickle cell formation due to HbA induction. In summary, 2'-MOE-SSOs are a promising therapy for forms of ß-thalassemia caused by mutations leading to aberrant splicing.

14.
J Transl Med ; 14: 255, 2016 09 02.
Article in English | MEDLINE | ID: mdl-27590532

ABSTRACT

BACKGROUND: Cellular biobanking is a key resource for collaborative networks planning to use same cells in studies aimed at solving a variety of biological and biomedical issues. This approach is of great importance in studies on ß-thalassemia, since the recruitment of patients and collection of specimens can represent a crucial and often limiting factor in the experimental planning. METHODS: Erythroid precursor cells were obtained from 72 patients, mostly ß-thalassemic, expanded and cryopreserved. Expression of globin genes was analyzed by real time RT-qPCR. Hemoglobin production was studied by HPLC. RESULTS: In this paper we describe the production and validation of a Thal-Biobank constituted by expanded erythroid precursor cells from ß-thalassemia patients. The biobanked samples were validated for maintenance of their phenotype after (a) cell isolation from same patients during independent phlebotomies, (b) freezing step in different biobanked cryovials, (c) thawing step and analysis at different time points. Reproducibility was confirmed by shipping the frozen biobanked cells to different laboratories, where the cells were thawed, cultured and analyzed using the same standardized procedures. The biobanked cells were stratified on the basis of their baseline level of fetal hemoglobin production and exposed to fetal hemoglobin inducers. CONCLUSION: The use of biobanked cells allows stratification of the patients with respect to fetal hemoglobin production and can be used for determining the response to the fetal hemoglobin inducer hydroxyurea and to gene therapy protocols with reproducible results.


Subject(s)
Biological Specimen Banks , beta-Thalassemia/pathology , Antigens, CD34/metabolism , Biomarkers/metabolism , Cell Differentiation/drug effects , Cells, Cultured , Chromatography, High Pressure Liquid , Cryopreservation , Erythroid Precursor Cells/drug effects , Erythroid Precursor Cells/metabolism , Erythropoietin/pharmacology , Fetal Hemoglobin/metabolism , Hemoglobins/genetics , Hemoglobins/metabolism , Humans , Kinetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reproducibility of Results
15.
Blood ; 128(8): 1139-43, 2016 08 25.
Article in English | MEDLINE | ID: mdl-27405777

ABSTRACT

Overcoming the silencing of the fetal γ-globin gene has been a long-standing goal in the treatment of sickle cell disease (SCD). The major transcriptional enhancer of the ß-globin locus, called the locus control region (LCR), dynamically interacts with the developmental stage-appropriate ß-type globin genes via chromatin looping, a process requiring the protein Ldb1. In adult erythroid cells, the LCR can be redirected from the adult ß- to the fetal γ-globin promoter by tethering Ldb1 to the human γ-globin promoter with custom-designed zinc finger (ZF) proteins (ZF-Ldb1), leading to reactivation of γ-globin gene expression. To compare this approach to pharmacologic reactivation of fetal hemoglobin (HbF), hematopoietic cells from patients with SCD were treated with a lentivirus expressing the ZF-Ldb1 or with chemical HbF inducers. The HbF increase in cells treated with ZF-Ldb1 was more than double that observed with decitabine and pomalidomide; butyrate had an intermediate effect whereas tranylcypromine and hydroxyurea showed relatively low HbF reactivation. ZF-Ldb1 showed comparatively little toxicity, and reduced sickle hemoglobin (HbS) synthesis as well as sickling of SCD erythroid cells under hypoxic conditions. The efficacy and low cytotoxicity of lentiviral-mediated ZF-Ldb1 gene transfer compared with the drug regimens support its therapeutic potential for the treatment of SCD.


Subject(s)
Anemia, Sickle Cell/metabolism , Chromatin/metabolism , Fetal Hemoglobin/metabolism , Adult , Antigens, CD34/metabolism , DNA-Binding Proteins , Erythroid Cells/metabolism , Hemoglobin, Sickle , Humans , LIM Domain Proteins , Transcription Factors , Zinc Fingers
16.
Biomed Res Int ; 2015: 687635, 2015.
Article in English | MEDLINE | ID: mdl-26097845

ABSTRACT

Mouse models that carry mutations causing thalassemia represent a suitable tool to test in vivo new mutation-specific therapeutic approaches. Transgenic mice carrying the ß-globin IVSI-6 mutation (the most frequent in Middle-Eastern regions and recurrent in Italy and Greece) are, at present, not available. We report the production and characterization of a transgenic mouse line (TG-ß-IVSI-6) carrying the IVSI-6 thalassemia point mutation within the human ß-globin gene. In the TG-ß-IVSI-6 mouse (a) the transgenic integration region is located in mouse chromosome 7; (b) the expression of the transgene is tissue specific; (c) as expected, normally spliced human ß-globin mRNA is produced, giving rise to ß-globin production and formation of a human-mouse tetrameric chimeric hemoglobin (mu) α-globin2/(hu) ß-globin2 and, more importantly, (d) the aberrant ß-globin-IVSI-6 RNAs are present in blood cells. The TG-ß-IVSI-6 mouse reproduces the molecular features of IVSI-6 ß-thalassemia and might be used as an in vivo model to characterize the effects of antisense oligodeoxynucleotides targeting the cryptic sites responsible for the generation of aberrantly spliced ß-globin RNA sequences, caused by the IVSI-6 mutation. These experiments are expected to be crucial for the development of a personalized therapy for ß-thalassemia.


Subject(s)
Mice, Transgenic , beta-Globins/genetics , beta-Thalassemia/genetics , Animals , Base Sequence , Greece , Hemoglobins/genetics , Humans , Italy , Mice , Phenotype , Point Mutation , RNA Splicing , beta-Thalassemia/pathology
17.
J Blood Med ; 6: 69-85, 2015.
Article in English | MEDLINE | ID: mdl-25737641

ABSTRACT

The ß-thalassemias are a group of hereditary hematological diseases caused by over 300 mutations of the adult ß-globin gene. Together with sickle cell anemia, thalassemia syndromes are among the most impactful diseases in developing countries, in which the lack of genetic counseling and prenatal diagnosis have contributed to the maintenance of a very high frequency of these genetic diseases in the population. Gene therapy for ß-thalassemia has recently seen steadily accelerating progress and has reached a crossroads in its development. Presently, data from past and ongoing clinical trials guide the design of further clinical and preclinical studies based on gene augmentation, while fundamental insights into globin switching and new technology developments have inspired the investigation of novel gene-therapy approaches. Moreover, human erythropoietic stem cells from ß-thalassemia patients have been the cellular targets of choice to date whereas future gene-therapy studies might increasingly draw on induced pluripotent stem cells. Herein, we summarize the most significant developments in ß-thalassemia gene therapy over the last decade, with a strong emphasis on the most recent findings, for ß-thalassemia model systems; for ß-, γ-, and anti-sickling ß-globin gene addition and combinatorial approaches including the latest results of clinical trials; and for novel approaches, such as transgene-mediated activation of γ-globin and genome editing using designer nucleases.

18.
Cell ; 158(4): 849-860, 2014 Aug 14.
Article in English | MEDLINE | ID: mdl-25126789

ABSTRACT

Distal enhancers commonly contact target promoters via chromatin looping. In erythroid cells, the locus control region (LCR) contacts ß-type globin genes in a developmental stage-specific manner to stimulate transcription. Previously, we induced LCR-promoter looping by tethering the self-association domain (SA) of Ldb1 to the ß-globin promoter via artificial zinc fingers. Here, we show that targeting the SA to a developmentally silenced embryonic globin gene in adult murine erythroblasts triggers its transcriptional reactivation. This activity depends on the LCR, consistent with an LCR-promoter looping mechanism. Strikingly, targeting the SA to the fetal γ-globin promoter in primary adult human erythroblasts increases γ-globin promoter-LCR contacts, stimulating transcription to approximately 85% of total ß-globin synthesis, with a reciprocal reduction in adult ß-globin expression. Our findings demonstrate that forced chromatin looping can override a stringent developmental gene expression program and suggest a novel approach to control the balance of globin gene transcription for therapeutic applications.


Subject(s)
Chromatin/metabolism , Fetal Hemoglobin/genetics , Genetic Techniques , Locus Control Region , Transcriptional Activation , beta-Globins/genetics , Animals , Antigens, CD34/metabolism , Chromatin/chemistry , Embryo, Mammalian/metabolism , Erythroblasts/metabolism , Hemoglobinopathies/genetics , Hemoglobinopathies/therapy , Humans , Mice , Primary Cell Culture
19.
Cancer Res ; 74(11): 3104-13, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24662918

ABSTRACT

Many of the gene mutations found in genetic disorders, including cancer, result in premature termination codons (PTC) and the rapid degradation of their mRNAs by nonsense-mediated RNA decay (NMD). We used virtual library screening, targeting a pocket in the SMG7 protein, a key component of the NMD mechanism, to identify compounds that disrupt the SMG7-UPF1 complex and inhibit NMD. Several of these compounds upregulated NMD-targeted mRNAs at nanomolar concentrations, with minimal toxicity in cell-based assays. As expected, pharmacologic NMD inhibition disrupted SMG7-UPF1 interactions. When used in cells with PTC-mutated p53, pharmacologic NMD inhibition combined with a PTC "read-through" drug led to restoration of full-length p53 protein, upregulation of p53 downstream transcripts, and cell death. These studies serve as proof-of-concept that pharmacologic NMD inhibitors can restore mRNA integrity in the presence of PTC and can be used as part of a strategy to restore full-length protein in a variety of genetic diseases.


Subject(s)
Codon, Nonsense , Nonsense Mediated mRNA Decay/drug effects , Small Molecule Libraries/pharmacology , Tumor Suppressor Protein p53/genetics , Carrier Proteins/genetics , Cell Death/drug effects , Cell Death/genetics , Cell Line , Cell Line, Tumor , HCT116 Cells , HEK293 Cells , HeLa Cells , Humans , Nonsense Mediated mRNA Decay/genetics , RNA Helicases , RNA, Messenger/genetics , Trans-Activators/genetics , Up-Regulation/drug effects
20.
Hematol Oncol Clin North Am ; 28(2): 375-86, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24589272

ABSTRACT

Use of new compound such as inhibitors of JAK2 or transforming growth factor ß-like molecules might soon revolutionize the treatment of ß-thalassemia and related disorders. However, this situation requires careful optimization, noting the potential for off-target immune suppression for JAK2 inhibitors and the lack of mechanistic insights for the use of the ligand trap soluble molecules that sequester ligands of activin receptor IIA and B.


Subject(s)
Erythrocytes/physiology , Erythropoiesis/physiology , Hemoglobinopathies/physiopathology , beta-Thalassemia/physiopathology , Activin Receptors, Type II/antagonists & inhibitors , Activin Receptors, Type II/metabolism , Erythrocytes/drug effects , Erythrocytes/metabolism , Erythropoiesis/drug effects , Hemoglobinopathies/drug therapy , Hemoglobinopathies/metabolism , Humans , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/metabolism , Molecular Targeted Therapy/methods , Molecular Targeted Therapy/trends , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/therapeutic use , beta-Thalassemia/drug therapy , beta-Thalassemia/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...