Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Nat Rev Genet ; 22(4): 216-234, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33303992

RESUMEN

Haematopoietic stem and progenitor cell (HSPC) gene therapy has emerged as an effective treatment modality for monogenic disorders of the blood system such as primary immunodeficiencies and ß-thalassaemia. Medicinal products based on autologous HSPCs corrected using lentiviral and gammaretroviral vectors have now been approved for clinical use, and the site-specific genome modification of HSPCs using gene editing techniques such as CRISPR-Cas9 has shown great clinical promise. Preclinical studies have shown engineered HSPCs could also be used to cross-correct non-haematopoietic cells in neurodegenerative metabolic diseases. Here, we review the most recent advances in HSPC gene therapy and discuss emerging strategies for using HSPC gene therapy for a range of diseases.


Asunto(s)
Terapia Genética , Trasplante de Células Madre Hematopoyéticas/métodos , Enfermedades de Inmunodeficiencia Primaria/terapia , Talasemia beta/terapia , Sistemas CRISPR-Cas/genética , Edición Génica/métodos , Células Madre Hematopoyéticas/citología , Humanos , Enfermedades de Inmunodeficiencia Primaria/genética , Talasemia beta/genética
2.
Blood ; 141(10): 1169-1179, 2023 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-36508706

RESUMEN

ß-Thalassemia (BT) is one of the most common genetic diseases worldwide and is caused by mutations affecting ß-globin production. The only curative treatment is allogenic hematopoietic stem/progenitor cells (HSPCs) transplantation, an approach limited by compatible donor availability and immunological complications. Therefore, transplantation of autologous, genetically-modified HSPCs is an attractive therapeutic option. However, current gene therapy strategies based on the use of lentiviral vectors are not equally effective in all patients and CRISPR/Cas9 nuclease-based strategies raise safety concerns. Thus, base editing strategies aiming to correct the genetic defect in patients' HSPCs could provide safe and effective treatment. Here, we developed a strategy to correct one of the most prevalent BT mutations (IVS1-110 [G>A]) using the SpRY-ABE8e base editor. RNA delivery of the base editing system was safe and led to ∼80% of gene correction in the HSPCs of patients with BT without causing dangerous double-strand DNA breaks. In HSPC-derived erythroid populations, this strategy was able to restore ß-globin production and correct inefficient erythropoiesis typically observed in BT both in vitro and in vivo. In conclusion, this proof-of-concept study paves the way for the development of a safe and effective autologous gene therapy approach for BT.


Asunto(s)
Talasemia beta , Humanos , Talasemia beta/genética , Talasemia beta/terapia , Edición Génica , Sistemas CRISPR-Cas , Mutación , Globinas beta/genética
3.
Mol Ther ; 32(1): 124-139, 2024 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-37990494

RESUMEN

Quiescent human hematopoietic stem cells (HSC) are ideal targets for gene therapy applications due to their preserved stemness and repopulation capacities; however, they have not been exploited extensively because of their resistance to genetic manipulation. We report here the development of a lentiviral transduction protocol that overcomes this resistance in long-term repopulating quiescent HSC, allowing their efficient genetic manipulation. Mechanistically, lentiviral vector transduction of quiescent HSC was found to be restricted at the level of vector entry and by limited pyrimidine pools. These restrictions were overcome by the combined addition of cyclosporin H (CsH) and deoxynucleosides (dNs) during lentiviral vector transduction. Clinically relevant transduction levels were paired with higher polyclonal engraftment of long-term repopulating HSC as compared with standard ex vivo cultured controls. These findings identify the cell-intrinsic barriers that restrict the transduction of quiescent HSC and provide a means to overcome them, paving the way for the genetic engineering of unstimulated HSC.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas , Humanos , Transducción Genética , Lentivirus/genética , Terapia Genética/métodos , Inmunidad Innata , Vectores Genéticos/genética , Antígenos CD34
4.
Blood ; 139(23): 3387-3401, 2022 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-35073399

RESUMEN

Rare hematopoietic stem and progenitor cell (HSPC) pools outside the bone marrow (BM) contribute to blood production in stress and disease but remain ill-defined. Although nonmobilized peripheral blood (PB) is routinely sampled for clinical management, the diagnosis and monitoring potential of PB HSPCs remain untapped, as no healthy PB HSPC baseline has been reported. Here we comprehensively delineate human extramedullary HSPC compartments comparing spleen, PB, and mobilized PB to BM using single-cell RNA-sequencing and/or functional assays. We uncovered HSPC features shared by extramedullary tissues and others unique to PB. First, in contrast to actively dividing BM HSPCs, we found no evidence of substantial ongoing hematopoiesis in extramedullary tissues at steady state but report increased splenic HSPC proliferative output during stress erythropoiesis. Second, extramedullary hematopoietic stem cells/multipotent progenitors (HSCs/MPPs) from spleen, PB, and mobilized PB share a common transcriptional signature and increased abundance of lineage-primed subsets compared with BM. Third, healthy PB HSPCs display a unique bias toward erythroid-megakaryocytic differentiation. At the HSC/MPP level, this is functionally imparted by a subset of phenotypic CD71+ HSCs/MPPs, exclusively producing erythrocytes and megakaryocytes, highly abundant in PB but rare in other adult tissues. Finally, the unique erythroid-megakaryocytic-skewing of PB is perturbed with age in essential thrombocythemia and ß-thalassemia. Collectively, we identify extramedullary lineage-primed HSPC reservoirs that are nonproliferative in situ and report involvement of splenic HSPCs during demand-adapted hematopoiesis. Our data also establish aberrant composition and function of circulating HSPCs as potential clinical indicators of BM dysfunction.


Asunto(s)
Hematopoyesis , Células Madre Hematopoyéticas , Adulto , Médula Ósea , Células de la Médula Ósea/fisiología , Eritropoyesis , Humanos , Megacariocitos
5.
Kidney Int ; 104(1): 61-73, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-36990212

RESUMEN

Anemia is a common complication of systemic inflammation. Proinflammatory cytokines both decrease erythroblast sensitivity to erythropoietin (EPO) and increase the levels of the hepatic hormone hepcidin, sequestering iron in stores and causing functional iron deficiency. Anemia of chronic kidney disease (CKD) is a peculiar form of anemia of inflammation, characterized by impaired EPO production paralleling progressive kidney damage. Traditional therapy based on increased EPO (often in combination with iron) may have off-target effects due to EPO interaction with its non-erythroid receptors. Transferrin Receptor 2 (Tfr2) is a mediator of the iron-erythropoiesis crosstalk. Its deletion in the liver hampers hepcidin production, increasing iron absorption, whereas its deletion in the hematopoietic compartment increases erythroid EPO sensitivity and red blood cell production. Here, we show that selective hematopoietic Tfr2 deletion ameliorates anemia in mice with sterile inflammation in the presence of normal kidney function, promoting EPO responsiveness and erythropoiesis without increasing serum EPO levels. In mice with CKD, characterized by absolute rather than functional iron deficiency, Tfr2 hematopoietic deletion had a similar effect on erythropoiesis but anemia improvement was transient because of limited iron availability. Also, increasing iron levels by downregulating only hepatic Tfr2 had a minor effect on anemia. However, simultaneous deletion of hematopoietic and hepatic Tfr2, stimulating erythropoiesis and increased iron supply, was sufficient to ameliorate anemia for the entire protocol. Thus, our results suggest that combined targeting of hematopoietic and hepatic Tfr2 may be a therapeutic option to balance erythropoiesis stimulation and iron increase, without affecting EPO levels.


Asunto(s)
Anemia , Eritropoyetina , Deficiencias de Hierro , Insuficiencia Renal Crónica , Ratones , Animales , Hierro/metabolismo , Eritropoyesis/genética , Hepcidinas/genética , Hepcidinas/metabolismo , Modelos Animales de Enfermedad , Anemia/etiología , Anemia/genética , Eritropoyetina/metabolismo , Inflamación/tratamiento farmacológico , Inflamación/complicaciones , Receptores de Transferrina/genética , Insuficiencia Renal Crónica/complicaciones , Insuficiencia Renal Crónica/tratamiento farmacológico , Insuficiencia Renal Crónica/genética
6.
Blood ; 136(5): 610-622, 2020 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-32344432

RESUMEN

Hematopoietic stem cells (HSCs) are regulated by signals from the bone marrow (BM) niche that tune hematopoiesis at steady state and in hematologic disorders. To understand HSC-niche interactions in altered nonmalignant homeostasis, we selected ß-thalassemia, a hemoglobin disorder, as a paradigm. In this severe congenital anemia, alterations secondary to the primary hemoglobin defect have a potential impact on HSC-niche cross talk. We report that HSCs in thalassemic mice (th3) have an impaired function, caused by the interaction with an altered BM niche. The HSC self-renewal defect is rescued after cell transplantation into a normal microenvironment, thus proving the active role of the BM stroma. Consistent with the common finding of osteoporosis in patients, we found reduced bone deposition with decreased levels of parathyroid hormone (PTH), which is a key regulator of bone metabolism but also of HSC activity. In vivo activation of PTH signaling through the reestablished Jagged1 and osteopontin levels correlated with the rescue of the functional pool of th3 HSCs by correcting HSC-niche cross talk. Reduced HSC quiescence was confirmed in thalassemic patients, along with altered features of the BM stromal niche. Our findings reveal a defect in HSCs in ß-thalassemia induced by an altered BM microenvironment and provide novel and relevant insight for improving transplantation and gene therapy approaches.


Asunto(s)
Médula Ósea/patología , Células Madre Hematopoyéticas/patología , Nicho de Células Madre , Talasemia beta/patología , Animales , Femenino , Hematopoyesis/fisiología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL
7.
Blood ; 136(17): 1968-1979, 2020 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-32556142

RESUMEN

ß-Thalassemia intermedia is a disorder characterized by ineffective erythropoiesis (IE), anemia, splenomegaly, and systemic iron overload. Novel approaches are being explored based on the modulation of pathways that reduce iron absorption (ie, using hepcidin activators like Tmprss6-antisense oligonucleotides [ASOs]) or increase erythropoiesis (by erythropoietin [EPO] administration or modulating the ability of transferrin receptor 2 [Tfr2] to control red blood cell [RBC] synthesis). Targeting Tmprss6 messenger RNA by Tmprss6-ASO was proven to be effective in improving IE and splenomegaly by inducing iron restriction. However, we postulated that combinatorial strategies might be superior to single therapies. Here, we combined Tmprss6-ASO with EPO administration or removal of a single Tfr2 allele in the bone marrow of animals affected by ß-thalassemia intermedia (Hbbth3/+). EPO administration alone or removal of a single Tfr2 allele increased hemoglobin levels and RBCs. However, EPO or Tfr2 single-allele deletion alone, respectively, exacerbated or did not improve splenomegaly in ß-thalassemic mice. To overcome this issue, we postulated that some level of iron restriction (by targeting Tmprss6) would improve splenomegaly while preserving the beneficial effects on RBC production mediated by EPO or Tfr2 deletion. While administration of Tmprss6-ASO alone improved the anemia, the combination of Tmprss6-ASO + EPO or Tmprss6-ASO + Tfr2 single-allele deletion produced significantly higher hemoglobin levels and reduced splenomegaly. In conclusion, our results clearly indicate that these combinatorial approaches are superior to single treatments in ameliorating IE and anemia in ß-thalassemia and could provide guidance to translate some of these approaches into viable therapies.


Asunto(s)
Eritropoyetina/administración & dosificación , Eritropoyetina/genética , Terapia Genética/métodos , Proteínas de la Membrana/antagonistas & inhibidores , Oligonucleótidos Antisentido/administración & dosificación , Talasemia beta/terapia , Animales , Células Cultivadas , Eritropoyesis/efectos de los fármacos , Eritropoyesis/genética , Regulación de la Expresión Génica/efectos de los fármacos , Hierro/metabolismo , Sobrecarga de Hierro/genética , Sobrecarga de Hierro/prevención & control , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Oligonucleótidos Antisentido/farmacología , Receptores de Transferrina/genética , Serina Endopeptidasas/genética , Serina Endopeptidasas/metabolismo , Talasemia beta/metabolismo
8.
Am J Hematol ; 97(10): 1324-1336, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36071579

RESUMEN

ß-thalassemia is a genetic disorder caused by mutations in the ß-globin gene, and characterized by anemia, ineffective erythropoiesis and iron overload. Patients affected by the most severe transfusion-dependent form of the disease (TDT) require lifelong blood transfusions and iron chelation therapy, a symptomatic treatment associated with several complications. Other therapeutic opportunities are available, but none is fully effective and/or applicable to all patients, calling for the identification of novel strategies. Transferrin receptor 2 (TFR2) balances red blood cells production according to iron availability, being an activator of the iron-regulatory hormone hepcidin in the liver and a modulator of erythropoietin signaling in erythroid cells. Selective Tfr2 deletion in the BM improves anemia and iron-overload in non-TDT mice, both as a monotherapy and, even more strikingly, in combination with iron-restricting approaches. However, whether Tfr2 targeting might represent a therapeutic option for TDT has never been investigated so far. Here, we prove that BM Tfr2 deletion improves anemia, erythrocytes morphology and ineffective erythropoiesis in the Hbbth1/th2 murine model of TDT. This effect is associated with a decrease in the expression of α-globin, which partially corrects the unbalance with ß-globin chains and limits the precipitation of misfolded hemoglobin, and with a decrease in the activation of unfolded protein response. Remarkably, BM Tfr2 deletion is also sufficient to avoid long-term blood transfusions required for survival of Hbbth1/th2 animals, preventing mortality due to chronic anemia and reducing transfusion-associated complications, such as progressive iron-loading. Altogether, TFR2 targeting might represent a promising therapeutic option also for TDT.


Asunto(s)
Sobrecarga de Hierro , Receptores de Transferrina , Talasemia beta , Animales , Transfusión Sanguínea , Modelos Animales de Enfermedad , Hierro/metabolismo , Sobrecarga de Hierro/genética , Sobrecarga de Hierro/metabolismo , Ratones , Receptores de Transferrina/genética , Globinas beta , Talasemia beta/genética , Talasemia beta/terapia
9.
Haematologica ; 106(3): 795-805, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-32107334

RESUMEN

Nuclear receptor coactivator 4 (NCOA4) promotes ferritin degradation and Ncoa4-ko mice in a C57BL/6 background show microcytosis and mild anemia, aggravated by iron deficiency. To understand tissue-specific contributions of NCOA4-mediated ferritinophagy we explored the effect of Ncoa4 genetic ablation in the iron-rich Sv129/J strain. Increased body iron content protects these mice from anemia and, in basal conditions, Sv129/J Ncoa4-ko mice show only microcytosis; nevertheless, when fed a low-iron diet they develop a more severe anemia compared to that of wild-type animals. Reciprocal bone marrow (BM) transplantation from wild-type donors into Ncoa4-ko and from Ncoa4-ko into wild-type mice revealed that microcytosis and susceptibility to iron deficiency anemia depend on BM-derived cells. Reconstitution of erythropoiesis with normalization of red blood count and hemoglobin concentration occurred at the same rate in transplanted animals independently of the genotype. Importantly, NCOA4 loss did not affect terminal erythropoiesis in iron deficiency, both in total and specific BM Ncoa4-ko animals compared to controls. On the contrary, upon a low iron diet, spleen from wild-type animals with Ncoa4-ko BM displayed marked iron retention compared to (wild-type BM) controls, indicating defective macrophage iron release in the former. Thus, erythropoietin administration failed to mobilize iron from stores in Ncoa4-ko animals. Furthermore, Ncoa4 inactivation in thalassemic mice did not worsen the hematologic phenotype. Overall our data reveal a major role for NCOA4-mediated ferritinophagy in macrophages to favor iron release for erythropoiesis, especially in iron deficiency.


Asunto(s)
Eritropoyesis , Coactivadores de Receptor Nuclear , Animales , Ferritinas , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Coactivadores de Receptor Nuclear/genética , Coactivadores de Receptor Nuclear/metabolismo
10.
Blood ; 132(21): 2286-2297, 2018 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-30209118

RESUMEN

ß-thalassemias are genetic disorders characterized by anemia, ineffective erythropoiesis, and iron overload. Current treatment of severe cases is based on blood transfusion and iron chelation or allogeneic bone marrow (BM) transplantation. Novel approaches are explored for nontransfusion-dependent patients (thalassemia intermedia) who develop anemia and iron overload. Here, we investigated the erythropoietin (EPO) receptor partner, transferrin receptor 2 (TFR2), as a novel potential therapeutic target. We generated a murine model of thalassemia intermedia specifically lacking BM Tfr2: because their erythroid cells are more susceptible to EPO stimulation, mice show improved erythropoiesis and red blood cell morphology as well as partial correction of anemia and iron overload. The beneficial effects become attenuated over time, possibly due to insufficient iron availability to sustain the enhanced erythropoiesis. Germ line deletion of Tfr2, including haploinsufficiency, had a similar effect in the thalassemic model. Because targeting TFR2 enhances EPO-mediated effects exclusively in cells expressing both receptors, this approach may have advantages over erythropoiesis-stimulating agents in the treatment of other anemias.


Asunto(s)
Anemia/genética , Eliminación de Gen , Sobrecarga de Hierro/genética , Receptores de Transferrina/genética , Talasemia beta/genética , Anemia/metabolismo , Anemia/patología , Anemia/terapia , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Células Eritroides/metabolismo , Células Eritroides/patología , Eritropoyesis , Eritropoyetina/metabolismo , Femenino , Terapia Genética , Sobrecarga de Hierro/metabolismo , Sobrecarga de Hierro/patología , Sobrecarga de Hierro/terapia , Masculino , Ratones Endogámicos C57BL , Receptores de Transferrina/metabolismo , Talasemia beta/metabolismo , Talasemia beta/patología , Talasemia beta/terapia
11.
Blood Cells Mol Dis ; 70: 87-101, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29336892

RESUMEN

Gene therapy for hemoglobinopathies is currently based on transplantation of autologous hematopoietic stem cells genetically modified with an integrating lentiviral vector expressing a globin gene under the control of globin transcriptional regulatory elements. Studies and safety works demonstrated the potential therapeutic efficacy and safety of this approach, providing the rationale for clinical translation. The outcomes of early clinical trials, although showing promising results, have highlighted the current limitations to a more general application. These include the nature, source and age of repopulating hematopoietic stem cells, the suboptimal transduction efficiency and gene expression levels, the toxicity and efficacy of bone marrow conditioning, the stress status of bone marrow microenvironment in chronic diseases such as ß-thalassemia and sickle cell disease. Recently, gene editing strategies based on the use of nucleases offered a novel approach to increase globin expression in a quasi-physiological way, independently from the addition of transgenes and viral sequences to the human genome. This review will discuss the current status of gene therapy for ß-thalassemia and sickle cell disease with a perspective towards the improvements necessary in the context of clinical translation.


Asunto(s)
Edición Génica , Terapia Genética , Hemoglobinopatías/genética , Hemoglobinopatías/terapia , Hemoglobinas/genética , Anemia de Células Falciformes/diagnóstico , Anemia de Células Falciformes/genética , Animales , Ensayos Clínicos como Asunto , Terapia Combinada , Predisposición Genética a la Enfermedad , Terapia Genética/métodos , Humanos , Resultado del Tratamiento , Talasemia beta/diagnóstico , Talasemia beta/genética
12.
Blood ; 125(7): 1170-9, 2015 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-25499454

RESUMEN

Transferrin receptor 2 (TFR2) contributes to hepcidin regulation in the liver and associates with erythropoietin receptor in erythroid cells. Nevertheless, TFR2 mutations cause iron overload (hemochromatosis type 3) without overt erythroid abnormalities. To clarify TFR2 erythroid function, we generated a mouse lacking Tfr2 exclusively in the bone marrow (Tfr2(BMKO)). Tfr2(BMKO) mice have normal iron parameters, reduced hepcidin levels, higher hemoglobin and red blood cell counts, and lower mean corpuscular volume than normal control mice, a phenotype that becomes more evident in iron deficiency. In Tfr2(BMKO) mice, the proportion of nucleated erythroid cells in the bone marrow is higher and the apoptosis lower than in controls, irrespective of comparable erythropoietin levels. Induction of moderate iron deficiency increases erythroblasts number, reduces apoptosis, and enhances erythropoietin (Epo) levels in controls, but not in Tfr2(BMKO) mice. Epo-target genes such as Bcl-xL and Epor are highly expressed in the spleen and in isolated erythroblasts from Tfr2(BMKO) mice. Low hepcidin expression in Tfr2(BMKO) is accounted for by erythroid expansion and production of the erythroid regulator erythroferrone. We suggest that Tfr2 is a component of a novel iron-sensing mechanism that adjusts erythrocyte production according to iron availability, likely by modulating the erythroblast Epo sensitivity.


Asunto(s)
Eritrocitos/fisiología , Eritropoyesis/genética , Receptores de Transferrina/fisiología , Animales , Apoptosis/genética , Recuento de Eritrocitos , Eritropoyetina/metabolismo , Femenino , Hemoglobinas/metabolismo , Hepcidinas/genética , Hepcidinas/metabolismo , Hierro/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
13.
Blood ; 119(21): 5021-9, 2012 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-22490684

RESUMEN

Inappropriately low expression of the key iron regulator hepcidin (HAMP) causes iron overload in untransfused patients affected by ß-thalassemia intermedia and Hamp modulation provides improvement of the thalassemic phenotype of the Hbb(th3/+) mouse. HAMP expression is activated by iron through the bone morphogenetic protein (BMP)-son of mothers against decapentaplegic signaling pathway and inhibited by ineffective erythropoiesis through an unknown "erythroid regulator." The BMP pathway is inactivated by the serine protease TMPRSS6 that cleaves the BMP coreceptor hemojuvelin. Here, we show that homozygous loss of Tmprss6 in Hbb(th3/+) mice improves anemia and reduces ineffective erythropoiesis, splenomegaly, and iron loading. All these effects are mediated by Hamp up-regulation, which inhibits iron absorption and recycling. Because Hbb(th3/+) mice lacking Tmprss6 show residual ineffective erythropoiesis, our results indicate that Tmprss6 is essential for Hamp inhibition by the erythroid regulator. We also obtained partial correction of the phenotype in Tmprss6 haploinsufficient Hbb(th3/+) male but not female mice and showed that the observed sex difference reflects an unequal balance between iron and erythropoiesis-mediated Hamp regulation. Our study indicates that preventing iron overload improves ß-thalassemia and strengthens the essential role of Tmprss6 for Hamp suppression, providing a proof of concept that Tmprss6 manipulation can offer a novel therapeutic option in this condition.


Asunto(s)
Eliminación de Gen , Sobrecarga de Hierro/genética , Proteínas de la Membrana/genética , Serina Endopeptidasas/genética , Talasemia beta/genética , Animales , Péptidos Catiónicos Antimicrobianos/antagonistas & inhibidores , Péptidos Catiónicos Antimicrobianos/genética , Péptidos Catiónicos Antimicrobianos/metabolismo , Modelos Animales de Enfermedad , Eritropoyesis/genética , Eritropoyesis/fisiología , Femenino , Hepcidinas , Sobrecarga de Hierro/etiología , Sobrecarga de Hierro/metabolismo , Sobrecarga de Hierro/prevención & control , Masculino , Proteínas de la Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Serina Endopeptidasas/fisiología , Regulación hacia Arriba/genética , Talasemia beta/complicaciones , Talasemia beta/metabolismo , Talasemia beta/patología
15.
Blood ; 117(13): 3669-79, 2011 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-21263153

RESUMEN

Sox6 belongs to the Sry (sex-determining region Y)-related high-mobility-group-box family of transcription factors, which control cell-fate specification of many cell types. Here, we explored the role of Sox6 in human erythropoiesis by its overexpression both in the erythroleukemic K562 cell line and in primary erythroid cultures from human cord blood CD34+ cells. Sox6 induced significant erythroid differentiation in both models. K562 cells underwent hemoglobinization and, despite their leukemic origin, died within 9 days after transduction; primary erythroid cultures accelerated their kinetics of erythroid maturation and increased the number of cells that reached the final enucleation step. Searching for direct Sox6 targets, we found SOCS3 (suppressor of cytokine signaling-3), a known mediator of cytokine response. Sox6 was bound in vitro and in vivo to an evolutionarily conserved regulatory SOCS3 element, which induced transcriptional activation. SOCS3 overexpression in K562 cells and in primary erythroid cells recapitulated the growth inhibition induced by Sox6, which demonstrates that SOCS3 is a relevant Sox6 effector.


Asunto(s)
Células Precursoras Eritroides/fisiología , Eritropoyesis/genética , Factores de Transcripción SOXD/fisiología , Animales , Antígenos CD34/metabolismo , Diferenciación Celular/genética , Procesos de Crecimiento Celular/genética , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Células Precursoras Eritroides/metabolismo , Eritropoyesis/fisiología , Regulación de la Expresión Génica/fisiología , Humanos , Células K562 , Ratones , Modelos Biológicos , Factores de Transcripción SOXD/genética , Factores de Transcripción SOXD/metabolismo , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/genética , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/fisiología , Transfección
16.
Nat Med ; 12(12): 1397-402, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17115047

RESUMEN

The continuous renewal of human epidermis is sustained by stem cells contained in the epidermal basal layer and in hair follicles. Cultured keratinocyte stem cells, known as holoclones, generate sheets of epithelium used to restore severe skin, mucosal and corneal defects. Mutations in genes encoding the basement membrane component laminin 5 (LAM5) cause junctional epidermolysis bullosa (JEB), a devastating and often fatal skin adhesion disorder. Epidermal stem cells from an adult patient affected by LAM5-beta3-deficient JEB were transduced with a retroviral vector expressing LAMB3 cDNA (encoding LAM5-beta3), and used to prepare genetically corrected cultured epidermal grafts. Nine grafts were transplanted onto surgically prepared regions of the patient's legs. Engraftment was complete after 8 d. Synthesis and proper assembly of normal levels of functional LAM5 were observed, together with the development of a firmly adherent epidermis that remained stable for the duration of the follow-up (1 year) in the absence of blisters, infections, inflammation or immune response. Retroviral integration site analysis indicated that the regenerated epidermis is maintained by a defined repertoire of transduced stem cells. These data show that ex vivo gene therapy of JEB is feasible and leads to full functional correction of the disease.


Asunto(s)
Células Epidérmicas , Epidermólisis Ampollosa de la Unión/terapia , Terapia Genética/métodos , Trasplante de Células Madre , Células 3T3 , Adulto , Animales , Moléculas de Adhesión Celular/genética , Células Cultivadas , Estudios de Factibilidad , Vectores Genéticos , Humanos , Masculino , Ratones , Retroviridae , Ingeniería de Tejidos/métodos , Kalinina
17.
Nucleic Acids Res ; 39(2): 486-501, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20852263

RESUMEN

The Sox6 transcription factor plays critical roles in various cell types, including erythroid cells. Sox6-deficient mice are anemic due to impaired red cell maturation and show inappropriate globin gene expression in definitive erythrocytes. To identify new Sox6 target genes in erythroid cells, we used the known repressive double Sox6 consensus within the εy-globin promoter to perform a bioinformatic genome-wide search for similar, evolutionarily conserved motifs located within genes whose expression changes during erythropoiesis. We found a highly conserved Sox6 consensus within the Sox6 human gene promoter itself. This sequence is bound by Sox6 in vitro and in vivo, and mediates transcriptional repression in transient transfections in human erythroleukemic K562 cells and in primary erythroblasts. The binding of a lentiviral transduced Sox6FLAG protein to the endogenous Sox6 promoter is accompanied, in erythroid cells, by strong downregulation of the endogenous Sox6 transcript and by decreased in vivo chromatin accessibility of this region to the PstI restriction enzyme. These observations suggest that the negative Sox6 autoregulation, mediated by the double Sox6 binding site within its own promoter, may be relevant to control the Sox6 transcriptional downregulation that we observe in human erythroid cultures and in mouse bone marrow cells in late erythroid maturation.


Asunto(s)
Regulación hacia Abajo/genética , Células Eritroides/metabolismo , Eritropoyesis/genética , Regiones Promotoras Genéticas , Factores de Transcripción SOXD/genética , Animales , Secuencia de Bases , Sitios de Unión , Células Cultivadas , Secuencia Conservada , Humanos , Células K562 , Ratones , Factores de Transcripción SOXD/metabolismo , Transcripción Genética
18.
Hum Gene Ther ; 34(17-18): 793-807, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37675899

RESUMEN

ß-Thalassemia and sickle cell disease are autosomal recessive disorders of red blood cells due to mutations in the adult ß-globin gene, with a worldwide diffusion. The severe forms of hemoglobinopathies are fatal if untreated, and allogeneic bone marrow transplantation can be offered to a limited proportion of patients. The unmet clinical need and the disease incidence have promoted the development of new genetic therapies based on the engineering of autologous hematopoietic stem cells. Here, the steps of ex vivo gene therapy development are reviewed along with results from clinical trials and recent new approaches employing cutting edge gene editing tools.


Asunto(s)
Anemia de Células Falciformes , Hemoglobinopatías , Talasemia beta , Adulto , Humanos , Hemoglobinopatías/genética , Hemoglobinopatías/terapia , Anemia de Células Falciformes/genética , Anemia de Células Falciformes/terapia , Talasemia beta/genética , Talasemia beta/terapia , Terapia Genética , Edición Génica
19.
Cell Stem Cell ; 30(5): 549-570, 2023 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-37146580

RESUMEN

The growing clinical success of hematopoietic stem/progenitor cell (HSPC) gene therapy (GT) relies on the development of viral vectors as portable "Trojan horses" for safe and efficient gene transfer. The recent advent of novel technologies enabling site-specific gene editing is broadening the scope and means of GT, paving the way to more precise genetic engineering and expanding the spectrum of diseases amenable to HSPC-GT. Here, we provide an overview of state-of-the-art and prospective developments of the HSPC-GT field, highlighting how advances in biological characterization and manipulation of HSPCs will enable the design of the next generation of these transforming therapeutics.


Asunto(s)
Sistemas CRISPR-Cas , Células Madre Hematopoyéticas , Estudios Prospectivos , Edición Génica , Terapia Genética , Biología
20.
Mol Ther Nucleic Acids ; 32: 229-246, 2023 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-37090420

RESUMEN

Sickle cell disease (SCD) is due to a mutation in the ß-globin gene causing production of the toxic sickle hemoglobin (HbS; α2ßS 2). Transplantation of autologous hematopoietic stem and progenitor cells (HSPCs) transduced with lentiviral vectors (LVs) expressing an anti-sickling ß-globin (ßAS) is a promising treatment; however, it is only partially effective, and patients still present elevated HbS levels. Here, we developed a bifunctional LV expressing ßAS3-globin and an artificial microRNA (amiRNA) specifically downregulating ßS-globin expression with the aim of reducing HbS levels and favoring ßAS3 incorporation into Hb tetramers. Efficient transduction of SCD HSPCs by the bifunctional LV led to a substantial decrease of ßS-globin transcripts in HSPC-derived erythroid cells, a significant reduction of HbS+ red cells, and effective correction of the sickling phenotype, outperforming ßAS gene addition and BCL11A gene silencing strategies. The bifunctional LV showed a standard integration profile, and neither HSPC viability, engraftment, and multilineage differentiation nor the erythroid transcriptome and miRNAome were affected by the treatment, confirming the safety of this therapeutic strategy. In conclusion, the combination of gene addition and gene silencing strategies can improve the efficacy of current LV-based therapeutic approaches without increasing the mutagenic vector load, thus representing a novel treatment for SCD.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA