Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
Cell ; 158(4): 849-860, 2014 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-25126789

RESUMO

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.


Assuntos
Cromatina/metabolismo , Hemoglobina Fetal/genética , Técnicas Genéticas , Região de Controle de Locus Gênico , Ativação Transcricional , Globinas beta/genética , Animais , Antígenos CD34/metabolismo , Cromatina/química , Embrião de Mamíferos/metabolismo , Eritroblastos/metabolismo , Hemoglobinopatias/genética , Hemoglobinopatias/terapia , Humanos , Camundongos , Cultura Primária de Células
2.
Mol Ther ; 29(4): 1625-1638, 2021 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-33515514

RESUMO

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.


Assuntos
Anemia Falciforme/genética , Transplante de Medula Óssea , Terapia Genética , Globinas beta/genética , Talassemia beta/genética , Anemia Falciforme/sangue , Anemia Falciforme/patologia , Anemia Falciforme/terapia , Animais , Expressão Gênica/genética , Vetores Genéticos/genética , Vetores Genéticos/farmacologia , Hemoglobinas/genética , Xenoenxertos , Humanos , Lentivirus/genética , Região de Controle de Locus Gênico/genética , Camundongos , Transdução Genética , Globinas beta/uso terapêutico , Talassemia beta/sangue , Talassemia beta/patologia , Talassemia beta/terapia
3.
Haematologica ; 106(5): 1433-1442, 2019 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-32439726

RESUMO

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.

4.
Anal Bioanal Chem ; 411(29): 7669-7680, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31273412

RESUMO

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.


Assuntos
Técnicas Biossensoriais , Diferenciação Celular/efeitos dos fármacos , Eritrócitos/efeitos dos fármacos , Ensaios de Triagem em Larga Escala/métodos , Regiões Promotoras Genéticas , Transcrição Gênica , Globinas beta/genética , gama-Globinas/genética , Eritrócitos/citologia , Hemoglobina Fetal/genética , Proteínas de Fluorescência Verde/genética , Humanos , Células K562 , Reprodutibilidade dos Testes
5.
Blood ; 128(8): 1139-43, 2016 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-27405777

RESUMO

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.


Assuntos
Anemia Falciforme/metabolismo , Cromatina/metabolismo , Hemoglobina Fetal/metabolismo , Adulto , Antígenos CD34/metabolismo , Proteínas de Ligação a DNA , Células Eritroides/metabolismo , Hemoglobina Falciforme , Humanos , Proteínas com Domínio LIM , Fatores de Transcrição , Dedos de Zinco
6.
J Transl Med ; 14: 255, 2016 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-27590532

RESUMO

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.


Assuntos
Bancos de Espécimes Biológicos , Talassemia beta/patologia , Antígenos CD34/metabolismo , Biomarcadores/metabolismo , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Cromatografia Líquida de Alta Pressão , Criopreservação , Células Precursoras Eritroides/efeitos dos fármacos , Células Precursoras Eritroides/metabolismo , Eritropoetina/farmacologia , Hemoglobina Fetal/metabolismo , Hemoglobinas/genética , Hemoglobinas/metabolismo , Humanos , Cinética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reprodutibilidade dos Testes
9.
bioRxiv ; 2024 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-38559013

RESUMO

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.

10.
Blood Adv ; 7(22): 6873-6885, 2023 11 28.
Artigo em Inglês | MEDLINE | ID: mdl-37672319

RESUMO

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.


Assuntos
Eritropoese , Talassemia beta , Humanos , Apoptose , Talassemia beta/genética , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Eritropoese/genética , Esplenomegalia
11.
Science ; 381(6656): 436-443, 2023 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-37499029

RESUMO

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.


Assuntos
Edição de Genes , Células-Tronco Hematopoéticas , Proteínas Proto-Oncogênicas c-kit , RNA Mensageiro , Edição de Genes/métodos , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/metabolismo , Proteínas Proto-Oncogênicas c-kit/genética , RNA Mensageiro/genética , Animais , Humanos , Camundongos
12.
Ann Hematol ; 91(8): 1201-13, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22460946

RESUMO

Gene therapy might fall short in achieving a complete reversion of the ß-thalassemic phenotype due to current limitations in vector design and myeloablative regimen. Following gene transfer, all or a large proportion of erythroid cells might express suboptimal levels of ß-globin, impairing the therapeutic potential of the treatment. Our aim was to evaluate whether, in absence of complete reversion of the ß-globin phenotype upon gene transfer, it is possible to use fetal hemoglobin induction to eliminate the residual α-globin aggregates and achieve normal levels of hemoglobin. Transgenic K562 cell lines and erythroid precursor cells from ß(0)39-thalassemia patients were employed. Gene therapy was performed with the lentiviral vector T9W. Induction of fetal hemoglobin was obtained using mithramycin. Levels of mRNA and hemoglobins were determined by qRT-PCR and HPLC. First, we analyzed the effect of mithramycin on K562 transgenic cell lines harboring different copies of a lentiviral vector carrying the human ß-globin gene, showing that γ-globin mRNA expression and HbF production can be induced in the presence of high levels of ß-globin gene expression and HbA accumulation. We then treated erythroid progenitor cells from ß-thalassemic patients with T9W, which expresses the human ß-globin gene and mithramycin separately or in combination. When transduction with our lentiviral vector is insufficient to completely eliminate the unpaired α-globin chains, combination of ß-globin gene transfer therapy together with fetal hemoglobin induction might be very efficacious to remove the excess of α-globin proteins in thalassemic erythroid progenitor cells.


Assuntos
Células Precursoras Eritroides/efeitos dos fármacos , Hemoglobina Fetal/metabolismo , Terapia Genética , Hemoglobina A/genética , Plicamicina/uso terapêutico , Talassemia beta/terapia , Adulto , Antibióticos Antineoplásicos/farmacologia , Antibióticos Antineoplásicos/uso terapêutico , Células Cultivadas , Terapia Combinada/métodos , Células Precursoras Eritroides/metabolismo , Células Precursoras Eritroides/fisiologia , Técnicas de Transferência de Genes , Terapia Genética/métodos , Células HEK293 , Hemoglobina A/metabolismo , Humanos , Células K562 , Plicamicina/farmacologia , Globinas beta/genética , Talassemia beta/genética , Talassemia beta/metabolismo
13.
Int J Paleopathol ; 33: 25-29, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33640560

RESUMO

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.


Assuntos
Holoprosencefalia , Cemitérios , Feto , Humanos , Masculino , Crânio/diagnóstico por imagem
14.
Leg Med (Tokyo) ; 48: 101801, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33268276

RESUMO

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.


Assuntos
Determinação da Idade pelos Dentes/métodos , Antropologia Forense/métodos , Dente Molar/anatomia & histologia , Germe de Dente/anatomia & histologia , Dente Decíduo/anatomia & histologia , Humanos , Recém-Nascido , Mandíbula , Maxila , Dente Molar/embriologia , Coroa do Dente/anatomia & histologia , Coroa do Dente/embriologia , Coroa do Dente/crescimento & desenvolvimento , Germe de Dente/embriologia , Dente Decíduo/embriologia
15.
Blood ; 112(3): 875-85, 2008 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-18480424

RESUMO

In beta-thalassemia, the mechanism driving ineffective erythropoiesis (IE) is insufficiently understood. We analyzed mice affected by beta-thalassemia and observed, unexpectedly, a relatively small increase in apoptosis of their erythroid cells compared with healthy mice. Therefore, we sought to determine whether IE could also be characterized by limited erythroid cell differentiation. In thalassemic mice, we observed that a greater than normal percentage of erythroid cells was in S-phase, exhibiting an erythroblast-like morphology. Thalassemic cells were associated with expression of cell cycle-promoting genes such as EpoR, Jak2, Cyclin-A, Cdk2, and Ki-67 and the antiapoptotic protein Bcl-X(L). The cells also differentiated less than normal erythroid ones in vitro. To investigate whether Jak2 could be responsible for the limited cell differentiation, we administered a Jak2 inhibitor, TG101209, to healthy and thalassemic mice. Exposure to TG101209 dramatically decreased the spleen size but also affected anemia. Although our data do not exclude a role for apoptosis in IE, we propose that expansion of the erythroid pool followed by limited cell differentiation exacerbates IE in thalassemia. In addition, these results suggest that use of Jak2 inhibitors has the potential to profoundly change the management of this disorder.


Assuntos
Diferenciação Celular , Células Eritroides/patologia , Eritropoese , Janus Quinase 2/genética , Talassemia beta/sangue , Animais , Apoptose , Quinases Ciclina-Dependentes/genética , Janus Quinase 2/antagonistas & inibidores , Camundongos , Baço/patologia
16.
Am J Hematol ; 84(11): 720-8, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19810011

RESUMO

In several types of thalassemia (including beta(0)39-thalassemia), stop codon mutations lead to premature translation termination and to mRNA destabilization through nonsense-mediated decay. Drugs (for instance aminoglycosides) can be designed to suppress premature termination, inducing a ribosomal readthrough. These findings have introduced new hopes for the development of a pharmacologic approach to the cure of this disease. However, the effects of aminoglycosides on globin mRNA carrying beta-thalassemia stop mutations have not yet been investigated. In this study, we have used a lentiviral construct containing the beta(0)39-thalassemia globin gene under control of the beta-globin promoter and a LCR cassette. We demonstrated by fluorescence-activated cell sorting (FACS) analysis the production of beta-globin by K562 cell clones expressing the beta(0)39-thalassemia globin gene and treated with G418. More importantly, after FACS and high-performance liquid chromatography (HPLC) analyses, erythroid precursor cells from beta(0)39-thalassemia patients were demonstrated to be able to produce beta-globin and adult hemoglobin after treatment with G418. This study strongly suggests that ribosomal readthrough should be considered a strategy for developing experimental strategies for the treatment of beta(0)-thalassemia caused by stop codon mutations. Am. J. Hematol., 2009. (c) 2009 Wiley-Liss, Inc.


Assuntos
Códon sem Sentido , Células Precursoras Eritroides/metabolismo , Gentamicinas/farmacologia , Hemoglobinas/biossíntese , Globinas beta/biossíntese , Talassemia beta/tratamento farmacológico , Células Cultivadas , Células Precursoras Eritroides/citologia , Hemoglobinas/genética , Homozigoto , Humanos , Células K562 , Globinas beta/efeitos dos fármacos , Globinas beta/genética , Talassemia beta/sangue , Talassemia beta/genética
17.
Biotechnol Appl Biochem ; 54(1): 41-52, 2009 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-19216718

RESUMO

Nonsense mutations, giving rise to UAA, UGA and UAG stop codons within the coding region of mRNAs, promote premature translational termination and are the leading cause of approx. 30% of inherited diseases, including cystic fibrosis, Duchenne muscular dystrophy and thalassaemia. For instance, in beta(0)39-thalassaemia the CAG (glutamine) codon is mutated to the UAG stop codon, leading to premature translation termination and to mRNA destabilization through the well-described NMD (nonsense-mediated mRNA decay). In order to develop an approach facilitating translation and, therefore, protection from NMD, aminoglycoside antibiotics have been tested on mRNAs carrying premature stop codons. These drugs decrease the accuracy in the codon-anticodon base-pairing, inducing a ribosomal read-through of the premature termination codons. Interestingly, recent papers have described drugs designed and produced for suppressing premature translational termination, inducing a ribosomal read-through of premature but not normal termination codons. These findings have introduced new hopes for the development of a pharmacological approach to the therapy of beta(0)39-thalassaemia. In this context, we started the development of a cellular model of the beta(0)39-thalassaemia mutation that could be used for the screening of a high number of aminoglycosides and analogous molecules. To this aim, we produced a lentiviral construct containing the beta(0)39-thalassaemia globin gene under a minimal LCR (locus control region) control and used this construct for the transduction of K562 cells, subsequently subcloned, with the purpose to obtain several K562 clones with different integration copies of the construct. These clones were then treated with Geneticin (also known as G418) and other aminoglycosides and the production of beta-globin was analysed by FACS analysis. The results obtained suggest that this experimental system is suitable for the characterization of correction of the beta(0)39-globin mutation causing beta-thalassaemia.


Assuntos
Códon sem Sentido , Células K562/fisiologia , Mutação Puntual , RNA Mensageiro/biossíntese , Globinas beta/genética , Talassemia beta/genética , Células Clonais , Clonagem Molecular , Expressão Gênica/efeitos dos fármacos , Gentamicinas/farmacologia , Proteínas de Fluorescência Verde/biossíntese , Proteínas de Fluorescência Verde/genética , Humanos , Lentivirus/genética , Mutagênese Sítio-Dirigida , Reação em Cadeia da Polimerase , RNA Mensageiro/genética , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/genética , Transcrição Gênica/efeitos dos fármacos , Transfecção , Globinas beta/biossíntese
18.
Mediterr J Hematol Infect Dis ; 11(1): e2019067, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31700592

RESUMO

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.

19.
Mol Diagn Ther ; 23(2): 173-186, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30701409

RESUMO

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.


Assuntos
Terapia Genética , Hemoglobinopatias/genética , Hemoglobinopatias/terapia , Globinas beta/genética , Epigênese Genética , Edição de Genes , Humanos , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA