Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
N Engl J Med ; 378(16): 1479-1493, 2018 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-29669226

RESUMO

BACKGROUND: Donor availability and transplantation-related risks limit the broad use of allogeneic hematopoietic-cell transplantation in patients with transfusion-dependent ß-thalassemia. After previously establishing that lentiviral transfer of a marked ß-globin (ßA-T87Q) gene could substitute for long-term red-cell transfusions in a patient with ß-thalassemia, we wanted to evaluate the safety and efficacy of such gene therapy in patients with transfusion-dependent ß-thalassemia. METHODS: In two phase 1-2 studies, we obtained mobilized autologous CD34+ cells from 22 patients (12 to 35 years of age) with transfusion-dependent ß-thalassemia and transduced the cells ex vivo with LentiGlobin BB305 vector, which encodes adult hemoglobin (HbA) with a T87Q amino acid substitution (HbAT87Q). The cells were then reinfused after the patients had undergone myeloablative busulfan conditioning. We subsequently monitored adverse events, vector integration, and levels of replication-competent lentivirus. Efficacy assessments included levels of total hemoglobin and HbAT87Q, transfusion requirements, and average vector copy number. RESULTS: At a median of 26 months (range, 15 to 42) after infusion of the gene-modified cells, all but 1 of the 13 patients who had a non-ß0/ß0 genotype had stopped receiving red-cell transfusions; the levels of HbAT87Q ranged from 3.4 to 10.0 g per deciliter, and the levels of total hemoglobin ranged from 8.2 to 13.7 g per deciliter. Correction of biologic markers of dyserythropoiesis was achieved in evaluated patients with hemoglobin levels near normal ranges. In 9 patients with a ß0/ß0 genotype or two copies of the IVS1-110 mutation, the median annualized transfusion volume was decreased by 73%, and red-cell transfusions were discontinued in 3 patients. Treatment-related adverse events were typical of those associated with autologous stem-cell transplantation. No clonal dominance related to vector integration was observed. CONCLUSIONS: Gene therapy with autologous CD34+ cells transduced with the BB305 vector reduced or eliminated the need for long-term red-cell transfusions in 22 patients with severe ß-thalassemia without serious adverse events related to the drug product. (Funded by Bluebird Bio and others; HGB-204 and HGB-205 ClinicalTrials.gov numbers, NCT01745120 and NCT02151526 .).


Assuntos
Terapia Genética , Globinas beta/genética , Talassemia beta/terapia , Adolescente , Adulto , Antígenos CD34 , Criança , Transfusão de Eritrócitos/estatística & dados numéricos , Feminino , Técnicas de Transferência de Genes , Vetores Genéticos , Hemoglobinas/análise , Hemoglobinas/genética , Humanos , Lentivirus/genética , Masculino , Mutação , Transplante Autólogo , Adulto Jovem , Talassemia beta/genética
2.
N Engl J Med ; 376(9): 848-855, 2017 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-28249145

RESUMO

Sickle cell disease results from a homozygous missense mutation in the ß-globin gene that causes polymerization of hemoglobin S. Gene therapy for patients with this disorder is complicated by the complex cellular abnormalities and challenges in achieving effective, persistent inhibition of polymerization of hemoglobin S. We describe our first patient treated with lentiviral vector-mediated addition of an antisickling ß-globin gene into autologous hematopoietic stem cells. Adverse events were consistent with busulfan conditioning. Fifteen months after treatment, the level of therapeutic antisickling ß-globin remained high (approximately 50% of ß-like-globin chains) without recurrence of sickle crises and with correction of the biologic hallmarks of the disease. (Funded by Bluebird Bio and others; HGB-205 ClinicalTrials.gov number, NCT02151526 .).


Assuntos
Anemia Falciforme/terapia , Terapia Genética , Globinas beta/genética , Adolescente , Anemia Falciforme/sangue , Ensaios Clínicos como Assunto , Expressão Gênica , Terapia Genética/efeitos adversos , Vetores Genéticos , Hemoglobina A/metabolismo , Humanos , Lentivirus , Masculino
3.
Hum Gene Ther ; 27(2): 148-65, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26886832

RESUMO

ß-globin gene disorders are the most prevalent inherited diseases worldwide and result from abnormal ß-globin synthesis or structure. Novel therapeutic approaches are being developed in an effort to move beyond palliative management. Gene therapy, by ex vivo lentiviral transfer of a therapeutic ß-globin gene derivative (ß(AT87Q)-globin) to hematopoietic stem cells, driven by cis-regulatory elements that confer high, erythroid-specific expression, has been evaluated in human clinical trials over the past 8 years. ß(AT87Q)-globin is used both as a strong inhibitor of HbS polymerization and as a biomarker. While long-term studies are underway in multiple centers in Europe and in the United States, proof-of-principle of efficacy and safety has already been obtained in multiple patients with ß-thalassemia and sickle cell disease.


Assuntos
Anemia Falciforme/terapia , Terapia Genética/métodos , Transplante de Células-Tronco Hematopoéticas , Globinas beta/genética , Talassemia beta/terapia , Anemia Falciforme/genética , Anemia Falciforme/metabolismo , Anemia Falciforme/patologia , Ensaios Clínicos como Assunto , Expressão Gênica , Técnicas de Transferência de Genes , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Células-Tronco Hematopoéticas , Humanos , Lentivirus/genética , Lentivirus/metabolismo , Segurança do Paciente , Transgenes , Condicionamento Pré-Transplante/métodos , Globinas beta/metabolismo , Talassemia beta/genética , Talassemia beta/metabolismo , Talassemia beta/patologia
4.
Curr Gene Ther ; 15(1): 64-81, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25429463

RESUMO

A previously published clinical trial demonstrated the benefit of autologous CD34(+) cells transduced with a selfinactivating lentiviral vector (HPV569) containing an engineered ß-globin gene (ß(A-T87Q)-globin) in a subject with ß thalassemia major. This vector has been modified to increase transduction efficacy without compromising safety. In vitro analyses indicated that the changes resulted in both increased vector titers (3 to 4 fold) and increased transduction efficacy (2 to 3 fold). An in vivo study in which 58 ß-thalassemic mice were transplanted with vector- or mock-transduced syngenic bone marrow cells indicated sustained therapeutic efficacy. Secondary transplantations involving 108 recipients were performed to evaluate long-term safety. The six month study showed no hematological or biochemical toxicity. Integration site (IS) profile revealed an oligo/polyclonal hematopoietic reconstitution in the primary transplants and reduced clonality in secondary transplants. Tumor cells were detected in the secondary transplant mice in all treatment groups (including the control group), without statistical differences in the tumor incidence. Immunohistochemistry and quantitative PCR demonstrated that tumor cells were not derived from transduced donor cells. This comprehensive efficacy and safety data provided the basis for initiating two clinical trials with this second generation vector (BB305) in Europe and in the USA in patients with ß-thalassemia major and sickle cell disease.


Assuntos
Anemia Falciforme/terapia , Terapia Genética/métodos , Vetores Genéticos , Lentivirus/genética , Talassemia beta/terapia , Anemia Falciforme/genética , Animais , Antígenos CD34/metabolismo , Biologia Computacional , Dano ao DNA , Modelos Animais de Doenças , Feminino , Expressão Gênica , Técnicas de Transferência de Genes , Transplante de Células-Tronco Hematopoéticas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Talassemia beta/genética
5.
Nature ; 514(7521): 242-6, 2014 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-25156257

RESUMO

ß-Thalassaemia major (ß-TM) is an inherited haemoglobinopathy caused by a quantitative defect in the synthesis of ß-globin chains of haemoglobin, leading to the accumulation of free α-globin chains that form toxic aggregates. Despite extensive knowledge of the molecular defects causing ß-TM, little is known of the mechanisms responsible for the ineffective erythropoiesis observed in the condition, which is characterized by accelerated erythroid differentiation, maturation arrest and apoptosis at the polychromatophilic stage. We have previously demonstrated that normal human erythroid maturation requires a transient activation of caspase-3 at the later stages of maturation. Although erythroid transcription factor GATA-1, the master transcriptional factor of erythropoiesis, is a caspase-3 target, it is not cleaved during erythroid differentiation. We have shown that, in human erythroblasts, the chaperone heat shock protein70 (HSP70) is constitutively expressed and, at later stages of maturation, translocates into the nucleus and protects GATA-1 from caspase-3 cleavage. The primary role of this ubiquitous chaperone is to participate in the refolding of proteins denatured by cytoplasmic stress, thus preventing their aggregation. Here we show in vitro that during the maturation of human ß-TM erythroblasts, HSP70 interacts directly with free α-globin chains. As a consequence, HSP70 is sequestrated in the cytoplasm and GATA-1 is no longer protected, resulting in end-stage maturation arrest and apoptosis. Transduction of a nuclear-targeted HSP70 mutant or a caspase-3-uncleavable GATA-1 mutant restores terminal maturation of ß-TM erythroblasts, which may provide a rationale for new targeted therapies of ß-TM.


Assuntos
Eritroblastos/metabolismo , Eritropoese , Proteínas de Choque Térmico HSP70/metabolismo , alfa-Globinas/metabolismo , Talassemia beta/sangue , Talassemia beta/metabolismo , Apoptose , Medula Óssea/metabolismo , Caspase 3/metabolismo , Núcleo Celular/metabolismo , Sobrevivência Celular/genética , Células Cultivadas , Citoplasma/metabolismo , Ativação Enzimática , Eritroblastos/citologia , Eritroblastos/patologia , Eritropoese/genética , Fator de Transcrição GATA1/genética , Fator de Transcrição GATA1/metabolismo , Regulação da Expressão Gênica , Proteínas de Choque Térmico HSP70/genética , Humanos , Cinética , Terapia de Alvo Molecular , Ligação Proteica , Redobramento de Proteína , Talassemia beta/patologia
6.
Nat Med ; 20(4): 398-407, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24658077

RESUMO

The pathophysiology of ineffective erythropoiesis in ß-thalassemia is poorly understood. We report that RAP-011, an activin receptor IIA (ActRIIA) ligand trap, improved ineffective erythropoiesis, corrected anemia and limited iron overload in a mouse model of ß-thalassemia intermedia. Expression of growth differentiation factor 11 (GDF11), an ActRIIA ligand, was increased in splenic erythroblasts from thalassemic mice and in erythroblasts and sera from subjects with ß-thalassemia. Inactivation of GDF11 decreased oxidative stress and the amount of α-globin membrane precipitates, resulting in increased terminal erythroid differentiation. Abnormal GDF11 expression was dependent on reactive oxygen species, suggesting the existence of an autocrine amplification loop in ß-thalassemia. GDF11 inactivation also corrected the abnormal ratio of immature/mature erythroblasts by inducing apoptosis of immature erythroblasts through the Fas-Fas ligand pathway. Taken together, these observations suggest that ActRIIA ligand traps may have therapeutic relevance in ß-thalassemia by suppressing the deleterious effects of GDF11, a cytokine which blocks terminal erythroid maturation through an autocrine amplification loop involving oxidative stress and α-globin precipitation.


Assuntos
Receptores de Activinas Tipo II/metabolismo , Proteínas Morfogenéticas Ósseas/antagonistas & inibidores , Eritroblastos/metabolismo , Eritropoese/efeitos dos fármacos , Fatores de Diferenciação de Crescimento/antagonistas & inibidores , Hematínicos/farmacologia , Proteínas Recombinantes de Fusão/farmacologia , Talassemia beta/metabolismo , Animais , Apoptose/fisiologia , Comunicação Autócrina/fisiologia , Proteínas Morfogenéticas Ósseas/metabolismo , Diferenciação Celular , Modelos Animais de Doenças , Proteína Ligante Fas , Amplificação de Genes/fisiologia , Fatores de Diferenciação de Crescimento/metabolismo , Ligantes , Camundongos , Estresse Oxidativo/fisiologia , Espécies Reativas de Oxigênio , Transdução de Sinais , Receptor fas
7.
Methods Enzymol ; 507: 109-24, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22365771

RESUMO

Patients with ß-thalassemia major require lifelong transfusions and iron chelation, regardless of the type of causative mutations (e.g., ß°, ß(E)/ß°). The only available curative therapy is allogeneic hematopoietic transplantation, although most patients do not have an HLA-matched, geno-identical donor, and those who do still risk graft-versus-host disease. Hence, gene therapy by ex vivo transfer of a functional ß-globin gene is an attractive novel therapeutic modality. In ß-thalassemia, transfer of a therapeutic globin gene does not confer a selective advantage to transduced stem cells, and complex DNA regulatory sequences have to be present within the transfer vector for proper expression. This is why lentiviral vectors have proven especially suited for this application, and the first Phase I/II human clinical trial was initiated. Here, we report on the first gene therapy patient with severe ß(E)/ß°-thalassemia, who has become transfusion-independent, and provide methods and protocols used in the context of this clinical trial.


Assuntos
Terapia Genética , Lentivirus/genética , Talassemia beta/terapia , Adolescente , Transplante de Medula Óssea , Técnicas de Cultura de Células , Células Cultivadas , Ensaios Clínicos como Assunto , Clonagem Molecular , Genes Virais , Engenharia Genética , Vetores Genéticos , Humanos , Lentivirus/isolamento & purificação , Masculino , Projetos de Pesquisa , Resultado do Tratamento
8.
Cell ; 145(3): 398-409, 2011 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-21529713

RESUMO

Sickle human hemoglobin (Hb) confers a survival advantage to individuals living in endemic areas of malaria, the disease caused by Plasmodium infection. As demonstrated hereby, mice expressing sickle Hb do not succumb to experimental cerebral malaria (ECM). This protective effect is exerted irrespectively of parasite load, revealing that sickle Hb confers host tolerance to Plasmodium infection. Sickle Hb induces the expression of heme oxygenase-1 (HO-1) in hematopoietic cells, via a mechanism involving the transcription factor NF-E2-related factor 2 (Nrf2). Carbon monoxide (CO), a byproduct of heme catabolism by HO-1, prevents further accumulation of circulating free heme after Plasmodium infection, suppressing the pathogenesis of ECM. Moreover, sickle Hb inhibits activation and/or expansion of pathogenic CD8(+) T cells recognizing antigens expressed by Plasmodium, an immunoregulatory effect that does not involve Nrf2 and/or HO-1. Our findings provide insight into molecular mechanisms via which sickle Hb confers host tolerance to severe forms of malaria.


Assuntos
Hemoglobina Falciforme/imunologia , Malária/imunologia , Plasmodium berghei , Animais , Linfócitos T CD8-Positivos/imunologia , Monóxido de Carbono/metabolismo , Quimiocinas/metabolismo , Cruzamentos Genéticos , Modelos Animais de Doenças , Heme Oxigenase-1/metabolismo , Interações Hospedeiro-Patógeno , Humanos , Malária/fisiopatologia , Malária Cerebral/imunologia , Malária Cerebral/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Fator 2 Relacionado a NF-E2/metabolismo
9.
Mol Ther ; 19(7): 1273-86, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21386821

RESUMO

A lentiviral vector encoding ß-globin flanked by insulator elements has been used to treat ß-thalassemia (ß-Thal) successfully in one human subject. However, a clonal expansion was observed after integration in the HMGA2 locus, raising the question of how commonly lentiviral integration would be associated with possible insertional activation. Here, we report correcting ß-Thal in a murine model using the same vector and a busulfan-conditioning regimen, allowing us to investigate efficacy and clonal evolution at 9.2 months after transplantation of bone marrow cells. The five gene-corrected recipient mice showed near normal levels of hemoglobin, reduced accumulation of reticulocytes, and normalization of spleen weights. Mapping of integration sites pretransplantation showed the expected favored integration in transcription units. The numbers of gene-corrected long-term repopulating cells deduced from the numbers of unique integrants indicated oligoclonal reconstitution. Clonal abundance was quantified using a Mu transposon-mediated method, indicating that clones with integration sites near growth-control genes were not enriched during growth. No integration sites involving HMGA2 were detected. Cells containing integration sites in genes became less common after prolonged growth, suggesting negative selection. Thus, ß-Thal gene correction in mice can be achieved without expansion of cells harboring vectors integrated near genes involved in growth control.


Assuntos
Vetores Genéticos/genética , Lentivirus/genética , Talassemia beta/terapia , Animais , Transplante de Medula Óssea , Cromatografia Líquida de Alta Pressão , Citometria de Fluxo , Proteína HMGA2/genética , Camundongos , Globinas beta/genética , Globinas beta/metabolismo , Talassemia beta/genética , Talassemia beta/metabolismo
10.
Blood ; 117(20): 5321-31, 2011 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-21436071

RESUMO

A challenge for gene therapy of genetic diseases is to maintain corrected cell populations in subjects undergoing transplantation in cases in which the corrected cells do not have intrinsic selective advantage over nontransduced cells. For inherited hematopoietic disorders, limitations include inefficient transduction of stem cell pools, the requirement for toxic myelosuppression, and a lack of optimal methods for cell selection after transduction. Here, we have designed a lentiviral vector that encodes human ß-globin and a truncated erythropoietin receptor, both under erythroid-specific transcriptional control. This truncated receptor confers enhanced sensitivity to erythropoietin and a benign course in human carriers. Transplantation of marrow transduced with the vector into syngenic thalassemic mice, which have elevated plasma erythropoietin levels, resulted in long-term correction of the disease even at low ratios of transduced/untransduced cells. Amplification of the red over the white blood cell lineages was self-controlled and averaged ∼ 100-fold instead of ∼ 5-fold for ß-globin expression alone. There was no detectable amplification of white blood cells or alteration of hematopoietic homeostasis. Notwithstanding legitimate safety concerns in the context of randomly integrating vectors, this approach may prove especially valuable in combination with targeted integration or in situ homologous recombination/repair and may lower the required level of pretransplantation myelosuppression.


Assuntos
Terapia Genética/métodos , Talassemia beta/terapia , Animais , Sequência de Bases , Primers do DNA/genética , Modelos Animais de Doenças , Eritropoese/genética , Expressão Gênica , Vetores Genéticos , Transplante de Células-Tronco Hematopoéticas , Homeostase , Humanos , Lentivirus/genética , Camundongos , Receptores da Eritropoetina/genética , Proteínas Recombinantes/genética , Transplante Isogênico , Globinas beta/genética , Talassemia beta/sangue , Talassemia beta/genética
11.
Nature ; 467(7313): 318-22, 2010 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-20844535

RESUMO

The ß-haemoglobinopathies are the most prevalent inherited disorders worldwide. Gene therapy of ß-thalassaemia is particularly challenging given the requirement for massive haemoglobin production in a lineage-specific manner and the lack of selective advantage for corrected haematopoietic stem cells. Compound ß(E)/ß(0)-thalassaemia is the most common form of severe thalassaemia in southeast Asian countries and their diasporas. The ß(E)-globin allele bears a point mutation that causes alternative splicing. The abnormally spliced form is non-coding, whereas the correctly spliced messenger RNA expresses a mutated ß(E)-globin with partial instability. When this is compounded with a non-functional ß(0) allele, a profound decrease in ß-globin synthesis results, and approximately half of ß(E)/ß(0)-thalassaemia patients are transfusion-dependent. The only available curative therapy is allogeneic haematopoietic stem cell transplantation, although most patients do not have a human-leukocyte-antigen-matched, geno-identical donor, and those who do still risk rejection or graft-versus-host disease. Here we show that, 33 months after lentiviral ß-globin gene transfer, an adult patient with severe ß(E)/ß(0)-thalassaemia dependent on monthly transfusions since early childhood has become transfusion independent for the past 21 months. Blood haemoglobin is maintained between 9 and 10 g dl(-1), of which one-third contains vector-encoded ß-globin. Most of the therapeutic benefit results from a dominant, myeloid-biased cell clone, in which the integrated vector causes transcriptional activation of HMGA2 in erythroid cells with further increased expression of a truncated HMGA2 mRNA insensitive to degradation by let-7 microRNAs. The clonal dominance that accompanies therapeutic efficacy may be coincidental and stochastic or result from a hitherto benign cell expansion caused by dysregulation of the HMGA2 gene in stem/progenitor cells.


Assuntos
Transfusão de Sangue , Terapia Genética , Proteína HMGA2/metabolismo , Globinas beta/genética , Globinas beta/metabolismo , Talassemia beta/genética , Talassemia beta/terapia , Adolescente , Células Sanguíneas/citologia , Células Sanguíneas/metabolismo , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Pré-Escolar , Células Clonais/metabolismo , Expressão Gênica , Vetores Genéticos/genética , Proteína HMGA2/genética , Homeostase , Humanos , Lentivirus/genética , Masculino , MicroRNAs/genética , Especificidade de Órgãos , RNA Mensageiro/análise , RNA Mensageiro/genética , Fatores de Tempo , Ativação Transcricional , Adulto Jovem , Talassemia beta/metabolismo
12.
FASEB J ; 22(6): 1849-60, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18245171

RESUMO

Pulmonary arterial hypertension (PAH) is one of the leading causes of morbidity and mortality in adult patients with sickle cell disease (SCD). Here, we developed a model to study the early stage of PAH in SCD. We exposed wild-type and transgenic sickle cell SAD (Hbb(s)/Hbb(s)) mice to hypoxia (8% O(2)) for 7 days. Prolonged hypoxia in SAD mice only induced 1) increased neutrophil count in both bronchoalveolar lavage (BAL) and peripheral circulation; 2) increased BAL IL1beta, IL10, IL6, and TNF-alpha; and 3) up-regulation of the genes endothelin-1, cyclo-oxygenase-2, angiotensin-converting-enzyme, and IL-1beta, suggesting that amplified inflammatory response and activation of the endothelin-1 system may contribute to the early phase of PAH in SCD. Since phosphodiesterases (PDEs) are involved in pulmonary vascular tone regulation, we evaluated gene expression of phosphodiesterase-4 (PDE-4) isoforms and of PDE-1, -2, -3, -7, -8, which are the main cyclic-adenosine-monophosphate hydrolyzing enzymes. In SAD mouse lungs, prolonged hypoxia significantly increased PDE-4 and -1 gene expressions. The PDE-4 inhibitor, rolipram, prevented the hypoxia-induced PDE-4 and -1 gene up-regulation and interfered with the development of PAH, most likely through modulation of both vascular tone and inflammatory factors. This finding supports a possible therapeutic use of PDEs inhibitors in the earlier phases of PAH in SCD.


Assuntos
Anemia Falciforme/complicações , Hipertensão Pulmonar/tratamento farmacológico , Inibidores da Fosfodiesterase 4 , Inibidores de Fosfodiesterase/farmacologia , Anemia Falciforme/tratamento farmacológico , Animais , Modelos Animais de Doenças , Hipóxia , Camundongos , Camundongos Transgênicos , Inibidores de Fosfodiesterase/uso terapêutico , Rolipram/farmacologia , Regulação para Cima/genética
13.
Exp Hematol ; 36(4): 412-23, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18295963

RESUMO

OBJECTIVE: To investigate whether expression of a membrane-anchored form of erythropoietin (MbEpo) results in self-controlled, autocrine proliferation, and differentiation of erythroid cells. This would provide a possible approach to the selective expansion of genetically corrected erythroid cells in gene-therapy protocols. MATERIALS AND METHODS: We designed retroviral vectors encoding MbEpo or secreted erythropoietin (Epo) and enhanced green fluorescent protein. Several Epo-dependent cell lines were transduced and their proliferative capacity evaluated. This approach was also assessed in human bone marrow CD34(+) cells and mouse bone marrow transplants. RESULTS: Retroviral vector-mediated MbEpo expression induced autocrine proliferation of the Epo-dependent cell lines DAE7 and UT7/Epo. However, it blocked the Epo receptor (EpoR)-induced activation of granulocyte macrophage colony-stimulating factor-dependent UT7/GM cells and the erythroid differentiation of both human hematopoietic cells in vitro and of mouse bone marrow cells in transplant experiments. MbEpo was present at the surface of UT7/GM cells. It did not affect the membrane localization of the EpoR, but prevented its normal Epo-dependent phosphorylation and internalization. By contrast to these inhibitory effects, a higher rate of EpoR replenishment in UT7/GM cells before MbEpo production rendered cell proliferation independent of exogenous growth factor. CONCLUSIONS: Activation of EpoR gene expression before MbEpo-induced EpoR activation is essential for activation or inhibition of growth and differentiation of Epo-dependent cell lines. It will be necessary to delay MbEpo expression in late erythroid progenitors until after EpoR gene activation, for erythroid cell expansion to be achieved in vivo.


Assuntos
Eritropoetina/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Receptores de Superfície Celular/metabolismo , Receptores da Eritropoetina/metabolismo , Animais , Transplante de Medula Óssea , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Eritropoetina/genética , Eritropoetina/farmacologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Ligação Proteica , Receptores de Superfície Celular/efeitos dos fármacos , Receptores da Eritropoetina/efeitos dos fármacos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Regulação para Cima
14.
PLoS Med ; 4(3): e90, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17388661

RESUMO

BACKGROUND: In breast cancers, only a minority of patients fully benefit from the different chemotherapy regimens currently in use. Identification of markers that could predict the response to a particular regimen would thus be critically important for patient care. In cell lines or animal models, tumor protein p53 (TP53) plays a critical role in modulating the response to genotoxic drugs. TP53 is activated in response to DNA damage and triggers either apoptosis or cell-cycle arrest, which have opposite effects on cell fate. Yet, studies linking TP53 status and chemotherapy response have so far failed to unambiguously establish this paradigm in patients. Breast cancers with a TP53 mutation were repeatedly shown to have a poor outcome, but whether this reflects poor response to treatment or greater intrinsic aggressiveness of the tumor is unknown. METHODS AND FINDINGS: In this study we analyzed 80 noninflammatory breast cancers treated by frontline (neoadjuvant) chemotherapy. Tumor diagnoses were performed on pretreatment biopsies, and the patients then received six cycles of a dose-dense regimen of 75 mg/m(2) epirubicin and 1,200 mg/m(2) cyclophosphamide, given every 14 days. After completion of chemotherapy, all patients underwent mastectomies, thus allowing for a reliable assessment of chemotherapy response. The pretreatment biopsy samples were used to determine the TP53 status through a highly efficient yeast functional assay and to perform RNA profiling. All 15 complete responses occurred among the 28 TP53-mutant tumors. Furthermore, among the TP53-mutant tumors, nine out of ten of the highly aggressive basal subtypes (defined by basal cytokeratin [KRT] immunohistochemical staining) experienced complete pathological responses, and only TP53 status and basal subtype were independent predictors of a complete response. Expression analysis identified many mutant TP53-associated genes, including CDC20, TTK, CDKN2A, and the stem cell gene PROM1, but failed to identify a transcriptional profile associated with complete responses among TP53 mutant tumors. In patients with unresponsive tumors, mutant TP53 status predicted significantly shorter overall survival. The 15 patients with responsive TP53-mutant tumors, however, had a favorable outcome, suggesting that this chemotherapy regimen can overcome the poor prognosis generally associated with mutant TP53 status. CONCLUSIONS: This study demonstrates that, in noninflammatory breast cancers, TP53 status is a key predictive factor for response to this dose-dense epirubicin-cyclophosphamide regimen and further suggests that the basal subtype is exquisitely sensitive to this association. Given the well-established predictive value of complete responses for long-term survival and the poor prognosis of basal and TP53-mutant tumors treated with other regimens, this chemotherapy could be particularly suited for breast cancer patients with a mutant TP53, particularly those with basal features.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Proteína Supressora de Tumor p53/genética , Ciclofosfamida/administração & dosagem , Epirubicina/administração & dosagem , Feminino , Perfilação da Expressão Gênica , Humanos , Pessoa de Meia-Idade , Dados de Sequência Molecular , Análise de Sequência com Séries de Oligonucleotídeos
15.
Blood ; 102(3): 1087-96, 2003 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-12689931

RESUMO

Central to the pathophysiology of sickle cell disease are the vaso-occlusive events that lead to tissue damages and life-threatening complications. Lungs are particularly vulnerable to vaso-occlusion because of their specific vasculature. We developed a mouse model of hypoxia/reoxygenation lung injury closely mimicking the lung pathology of patients with sickle cell disease. This model involves the exposure of transgenic sickle cell (SAD) mice to hypoxia (8% oxygen) for 4, 10, and 46 hours followed by 2 hours of reoxygenation. Gene expression profiling of SAD lung tissue pointed to the specific induction of genes involved in the response to ischemic stress and microcirculation remodeling: Hspcb, Hsp86-1, Nfe2l2, Ace, and Fgf7. Hypoxia/reoxygenation also induced a marked increase in bronchoalveolar (BAL) total leukocyte and neutrophil counts, BAL total protein content, and BAL tumor necrosis factor alpha (TNF-alpha), interleukin 6 (IL-6), IL-1alpha, and macrophage inflammatory protein 2 (MIP-2) levels, all indicators of enhanced inflammatory response as compared with control mice. Nitric oxide (NO) was administered to SAD mice. NO (40 ppm) inhalation protected SAD mice from the histopathologic lesions of ischemic/reperfusion lung injury with corresponding normalization and/or modulation of tissue gene expression profiles. Inhaled NO (1) significantly reduced the increase in BAL total protein content, BAL total leukocyte, and neutrophil counts; (2) modulated BAL cytokine network; and (3) did not affect hemoglobin and methemoglobin levels. The present study provides evidences for the beneficial effects of inhaled NO in pulmonary injury induced by hypoxia/reoxygenation in a mouse model of sickle cell disease (SCD) and opens new avenues in drug design based on tissue gene expression profiling.


Assuntos
Anemia Falciforme/complicações , Hipóxia/tratamento farmacológico , Óxido Nítrico/farmacologia , Pneumopatia Veno-Oclusiva/tratamento farmacológico , Administração por Inalação , Anemia Falciforme/tratamento farmacológico , Anemia Falciforme/patologia , Animais , Líquido da Lavagem Broncoalveolar/química , Líquido da Lavagem Broncoalveolar/citologia , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Hipóxia/patologia , Pulmão/patologia , Camundongos , Camundongos Transgênicos , Óxido Nítrico/administração & dosagem , Oxigênio/metabolismo , Pneumopatia Veno-Oclusiva/patologia
16.
Proc Natl Acad Sci U S A ; 99(22): 14380-5, 2002 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-12391330

RESUMO

Achieving long-term pancellular expression of a transferred gene at therapeutic level in a given hematopoietic lineage remains an important goal of gene therapy. Advances have recently been made in the genetic correction of the hemoglobinopathies by means of lentiviral vectors and large locus control region (LCR) derivatives. However, panerythroid beta globin gene expression has not yet been achieved in beta thalassemic mice because of incomplete transduction of the hematopoietic stem cell compartment and position effect variegation of proviruses integrated at a single copy per genome. Here, we report the permanent, panerythroid correction of severe beta thalassemia in mice, resulting from a homozygous deletion of the beta major globin gene, by transplantation of syngeneic bone marrow transduced with an HIV-1-derived [beta globin gene/LCR] lentiviral vector also containing the Rev responsive element and the central polypurine tract/DNA flap. The viral titers produced were high enough to achieve transduction of virtually all of the hematopoietic stem cells in the graft with an average of three integrated proviral copies per genome in all transplanted mice; the transduction was sustained for >7 months in both primary and secondary transplants, at which time approximately 95% of the red blood cells in all mice contained human beta globin contributing to 32 +/- 4% of all beta-like globin chains. Hematological parameters approached complete phenotypic correction, as assessed by hemoglobin levels and reticulocyte and red blood cell counts. All circulating red blood cells became and remained normocytic and normochromic, and their density was normalized. Free alpha globin chains were completely cleared from red blood cell membranes, splenomegaly abated, and iron deposit was almost eliminated in liver sections. These findings indicate that virtually complete transduction of the hematopoietic stem cell compartment can be achieved by high-titer lentiviral vectors and that position effect variegation can be mitigated by multiple events of proviral integration to yield balanced, panerythroid expression. These results provide a solid foundation for the initiation of human clinical trials in beta thalassemia patients.


Assuntos
Células Precursoras Eritroides/metabolismo , Vetores Genéticos , Globinas/genética , Lentivirus , Integração Viral , Talassemia beta/terapia , Células 3T3 , Animais , Células COS , Chlorocebus aethiops , Modelos Animais de Doenças , Eritrócitos/citologia , Expressão Gênica , Vetores Genéticos/genética , Humanos , Lentivirus/genética , Fígado/patologia , Camundongos , Camundongos Endogâmicos C57BL , Baço/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA