Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Haematologica ; 108(10): 2652-2663, 2023 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-37021532

RESUMEN

Clinical trials have shown that lentiviral-mediated gene therapy can ameliorate bone marrow failure (BMF) in nonconditioned Fanconi anemia (FA) patients resulting from the proliferative advantage of corrected FA hematopoietic stem and progenitor cells (HSPC). However, it is not yet known if gene therapy can revert affected molecular pathways in diseased HSPC. Single-cell RNA sequencing was performed in chimeric populations of corrected and uncorrected HSPC co-existing in the BM of gene therapy-treated FA patients. Our study demonstrates that gene therapy reverts the transcriptional signature of FA HSPC, which then resemble the transcriptional program of healthy donor HSPC. This includes a down-regulated expression of TGF-ß and p21, typically up-regulated in FA HSPC, and upregulation of DNA damage response and telomere maintenance pathways. Our results show for the first time the potential of gene therapy to rescue defects in the HSPC transcriptional program from patients with inherited diseases; in this case, in FA characterized by BMF and cancer predisposition.


Asunto(s)
Anemia de Fanconi , Pancitopenia , Humanos , Anemia de Fanconi/genética , Anemia de Fanconi/terapia , Anemia de Fanconi/metabolismo , Células Madre Hematopoyéticas/metabolismo , Terapia Genética/métodos , Factor de Crecimiento Transformador beta/metabolismo , Regulación hacia Arriba , Pancitopenia/metabolismo , Trastornos de Fallo de la Médula Ósea/metabolismo
2.
Cytotherapy ; 25(3): 277-285, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36610813

RESUMEN

The clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 system has revolutionized the gene editing field, making it possible to interrupt, insert or replace a sequence of interest with high precision in the human genome. Its easy design and wide applicability open up a variety of therapeutic alternatives for the treatment of genetic diseases. Indeed, very promising approaches for the correction of hematological disorders have been developed in the recent years, based on the self-renewal and multipotent differentiation properties of hematopoietic stem and progenitor cells, which make this cell subset the ideal target for gene therapy purposes. This technology has been applied in different congenital blood disorders, such as primary immunodeficiencies, X-linked severe combined immunodeficiency, X-linked chronic granulomatous disease or Wiskott-Aldrich syndrome, and inherited bone marrow failure syndromes, such as Fanconi anemia, congenital amegakaryocytic thrombocytopenia or severe congenital neutropenia. Furthermore, CRISPR/Cas9-based gene editing has been implemented successfully as a novel therapy for cancer immunotherapy, by the development of promising strategies such as the use of oncolytic viruses or adoptive cellular therapy to the chimeric antigen receptor-T-cell therapy. Therefore, considering the variety of genes and mutations affected, we can take advantage of the different DNA repair mechanisms by CRISPR/Cas9 in different manners, from homology-directed repair to non-homologous-end-joining to the latest emerging technologies such as base and prime editing. Although the delivery systems into hematopoietic stem and progenitor cells are still the bottleneck of this technology, some of the advances in genome editing shown in this review have already reached a clinical stage and show very promising preliminary results.


Asunto(s)
Sistemas CRISPR-Cas , Edición Génica , Humanos , Edición Génica/métodos , Sistemas CRISPR-Cas/genética , Terapia Genética/métodos , Mutación
3.
Pancreatology ; 21(2): 466-472, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33454209

RESUMEN

INTRODUCTION: Postoperative pancreatic fistula (POPF) is the most dreadful complication of pancreaticoduodenectomy (PD) and previous literature focused on technical modifications of pancreatic remnant reconstruction. We developed a multifactorial mitigation strategy (MS) and the aim of the study is to assess its clinical impact in patients at high-risk of POPF. METHODS: All patients candidate to PD between 2012 and 2018 were considered. Only patients with a high Fistula Risk Score (FRS 7-10) were included. Patients undergoing MS were compared to patients receiving Standard Strategy (SS). Clinical outcomes were compared between the two groups. Multivariate hierarchical logistic regression analyses were performed to detect independent predictors of POPF. RESULTS: Out of 212 patients, 33 were finally included in MS Group and 29 in SS Group. POPF rate was significantly lower in MS Group (12.1% vs 44.8%, p = 0.005). Delayed gastric emptying, postoperative pancreatitis, complications and hospital stay were also significantly lower in MS Group. Hierarchical logistic regression analyses showed that Body Mass Index (OR = 1.196, p = 0.036) and MS (OR = 0.187, p = 0.032) were independently associated with POPF. CONCLUSION: A multifactorial MS can be helpful to reduce POPF rate in patients with high FRS following PD. Personalized approach for vulnerable patients should be investigated in the future.


Asunto(s)
Fístula Pancreática/prevención & control , Pancreaticoduodenectomía/efectos adversos , Pancreatoyeyunostomía/efectos adversos , Complicaciones Posoperatorias/prevención & control , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Masculino , Persona de Mediana Edad , Fístula Pancreática/etiología , Factores de Riesgo
4.
Am J Hematol ; 96(8): 989-999, 2021 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-33984160

RESUMEN

Fanconi anemia (FA) is characterized by chromosome fragility, bone marrow failure (BMF) and predisposition to cancer. As reverse genetic mosaicism has been described as "natural gene therapy" in patients with FA, we sought to evaluate the clinical course of a cohort of FA mosaic patients followed at referral centers in Spain over a 30-year period. This cohort includes patients with a majority of T cells without chromosomal aberrations in the DEB-chromosomal breakage test. Relative to non-mosaic FA patients, we observed a higher proportion of adult patients in the cohort of mosaics, with a later age of hematologic onset and a milder evolution of (BMF). Consequently, the requirement for hematopoietic stem cell transplant (HSCT) was also lower. Additional studies allowed us to identify a sub-cohort of mosaic FA patients in whom the reversion was present in bone marrow (BM) progenitor cells leading to multilineage mosaicism. These multilineage mosaic patients are older, have a lower percentage of aberrant cells, have more stable hematology and none of them developed leukemia or myelodysplastic syndrome when compared to non-mosaics. In conclusion, our data indicate that reverse mosaicism is a good prognostic factor in FA and is associated with more favorable long-term clinical outcomes.


Asunto(s)
Anemia de Fanconi/terapia , Terapia Genética/métodos , Adolescente , Adulto , Niño , Anemia de Fanconi/genética , Humanos , Masculino , Mosaicismo , Adulto Joven
5.
J Med Genet ; 57(4): 258-268, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31586946

RESUMEN

PURPOSE: Patients with Fanconi anaemia (FA), a rare DNA repair genetic disease, exhibit chromosome fragility, bone marrow failure, malformations and cancer susceptibility. FA molecular diagnosis is challenging since FA is caused by point mutations and large deletions in 22 genes following three heritability patterns. To optimise FA patients' characterisation, we developed a simplified but effective methodology based on whole exome sequencing (WES) and functional studies. METHODS: 68 patients with FA were analysed by commercial WES services. Copy number variations were evaluated by sequencing data analysis with RStudio. To test FANCA missense variants, wt FANCA cDNA was cloned and variants were introduced by site-directed mutagenesis. Vectors were then tested for their ability to complement DNA repair defects of a FANCA-KO human cell line generated by TALEN technologies. RESULTS: We identified 93.3% of mutated alleles including large deletions. We determined the pathogenicity of three FANCA missense variants and demonstrated that two FANCA variants reported in mutations databases as 'affecting functions' are SNPs. Deep analysis of sequencing data revealed patients' true mutations, highlighting the importance of functional analysis. In one patient, no pathogenic variant could be identified in any of the 22 known FA genes, and in seven patients, only one deleterious variant could be identified (three patients each with FANCA and FANCD2 and one patient with FANCE mutations) CONCLUSION: WES and proper bioinformatics analysis are sufficient to effectively characterise patients with FA regardless of the rarity of their complementation group, type of mutations, mosaic condition and DNA source.


Asunto(s)
Secuenciación del Exoma , Proteína del Grupo de Complementación A de la Anemia de Fanconi/genética , Anemia de Fanconi/genética , Predisposición Genética a la Enfermedad , Línea Celular , Variaciones en el Número de Copia de ADN/genética , Reparación del ADN/genética , Proteínas de Unión al ADN/genética , Anemia de Fanconi/patología , Femenino , Técnicas de Inactivación de Genes , Humanos , Masculino , Mutación Missense/genética , Polimorfismo de Nucleótido Simple/genética
6.
Langenbecks Arch Surg ; 406(5): 1443-1452, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-33475833

RESUMEN

INTRODUCTION: The NCCN classification of resectability in pancreatic head cancer does not consider preoperative radiological tumour ≤ 180° contact with portal vein/superior mesenteric vein (PV/SMV) as a negative prognostic feature. The aim of this study is to evaluate whether this factor is associated with higher rate of incomplete resection and poorer survival. METHODS: All patients considered for pancreatic resection between 2012 and 2017 at two Spanish referral centres were included. Patients with borderline and locally advanced pancreatic ductal adenocarcinoma (PDAC) according to NCCN classification were excluded. Preoperative CT scans were reviewed by dedicated radiologists to identify radiologic tumour contact with PV/SMV. RESULTS: Out of 302, 71 patients were finally included in this study. Twenty-two (31%) patients showed tumour-PV/SMV contact (group 1) and 49 (69%) did not show any contact (group 2). Patients in group 1 showed a statistically significantly higher rate of R1 and R1-direct margins compared with group 2 (95 vs 28% and 77 vs 10%) and lower median survival (24 vs 41 months, p = 0.02). Preoperative contact with PV/SMV, lymph node metastases, R1-direct margin and NO adjuvant chemotherapy were significantly associated with disease-specific survival at multivariate analysis. CONCLUSION: Preoperative radiological tumour contact with PV/SMV in patients with NCCN resectable PDAC is associated with high rate of pathologic positive margins following surgery and poorer survival.


Asunto(s)
Venas Mesentéricas , Neoplasias Pancreáticas , Humanos , Venas Mesentéricas/diagnóstico por imagen , Venas Mesentéricas/cirugía , Invasividad Neoplásica , Neoplasias Pancreáticas/diagnóstico por imagen , Neoplasias Pancreáticas/cirugía , Pancreaticoduodenectomía , Vena Porta/diagnóstico por imagen , Vena Porta/cirugía , Estudios Retrospectivos
7.
Clin Genet ; 97(1): 89-102, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31231794

RESUMEN

Hematopoietic gene therapy has markedly progressed during the last 15 years both in terms of safety and efficacy. While a number of serious adverse events (SAE) were initially generated as a consequence of genotoxic insertions of gamma-retroviral vectors in the cell genome, no SAEs and excellent outcomes have been reported in patients infused with autologous hematopoietic stem cells (HSCs) transduced with self-inactivated lentiviral and gammaretroviral vectors. Advances in the field of HSC gene therapy have extended the number of monogenic diseases that can be treated with these approaches. Nowadays, evidence of clinical efficacy has been shown not only in primary immunodeficiencies, but also in other hematopoietic diseases, including beta-thalassemia and sickle cell anemia. In addition to the rapid progression of non-targeted gene therapies in the clinic, new approaches based on gene editing have been developed thanks to the discovery of designed nucleases and improved non-integrative vectors, which have markedly increased the efficacy and specificity of gene targeting to levels compatible with its clinical application. Based on advances achieved in the field of gene therapy, it can be envisaged that these therapies will soon be part of the therapeutic approaches used to treat life-threatening diseases of the hematopoietic system.


Asunto(s)
Anemia de Células Falciformes/terapia , Terapia Genética/tendencias , Enfermedades Hematológicas/terapia , Talasemia beta/terapia , Anemia de Células Falciformes/sangre , Células Sanguíneas/patología , Células Sanguíneas/trasplante , Vectores Genéticos/efectos adversos , Enfermedades Hematológicas/sangre , Enfermedades Hematológicas/patología , Trasplante de Células Madre Hematopoyéticas/tendencias , Células Madre Hematopoyéticas/citología , Humanos , Talasemia beta/sangre
8.
Ann Hematol ; 99(5): 913-924, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32065290

RESUMEN

Fanconi anemia (FA) is a DNA repair disorder resulting from mutations in genes encoding for FA DNA repair complex components and is characterized by variable congenital abnormalities, bone marrow failure (BMF), and high incidences of malignancies. FA mosaicism arises from reversion or other compensatory mutations in hematopoietic cells and may be associated with BMF reversal and decreased blood cell sensitivity to DNA-damaging agents (clastogens); this sensitivity is a phenotypic and diagnostic hallmark of FA. Uncertainty regarding the clinical significance of FA mosaicism persists; in some cases, patients have survived multiple decades without BMF or hematologic malignancy, and in others hematologic failure occurred despite the presence of clastogen-resistant cell populations. Assessment of mosaicism is further complicated because clinical evaluation is frequently based on clastogen resistance in lymphocytes, which may arise from reversion events both in lymphoid-specific lineages and in more pluripotent hematopoietic stem/progenitor cells (HSPCs). In this review, we describe diagnostic methods and outcomes in published mosaicism series, including the substantial intervals (1-6 years) over which blood counts normalized, and the relatively favorable clinical course in cases where clastogen resistance was demonstrated in bone marrow progenitors. We also analyzed published FA mosaic cases with emphasis on long-term clinical outcomes when blood count normalization was identified. Blood count normalization in FA mosaicism likely arises from reversion events in long-term primitive HSPCs and is associated with low incidences of BMF or hematologic malignancy. These observations have ramifications for current investigational therapeutic programs in FA intended to enable gene correction in long-term repopulating HSPCs.


Asunto(s)
Células de la Médula Ósea/metabolismo , Anemia de Fanconi , Neoplasias Hematológicas , Células Madre Hematopoyéticas/metabolismo , Mosaicismo , Células de la Médula Ósea/patología , Anemia de Fanconi/sangre , Anemia de Fanconi/diagnóstico , Anemia de Fanconi/genética , Anemia de Fanconi/terapia , Neoplasias Hematológicas/sangre , Neoplasias Hematológicas/diagnóstico , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/terapia , Humanos
9.
Ann Surg ; 270(5): 738-746, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31498183

RESUMEN

OBJECTIVE: To compare the rates of R0 resection in pancreatoduodenectomy (PD) for pancreatic and periampullary malignant tumors by means of standard (ST-PD) versus artery-first approach (AFA-PD). BACKGROUND: Standardized histological examination of PD specimens has shown that most pancreatic resections thought to be R0 resections are R1. "Artery-first approach" is a surgical technique characterized by meticulous dissection of arterial planes and clearing of retropancreatic tissue in an attempt to achieve a higher rate of R0. To date, studies comparing AFA-PD versus ST-PD are retrospective cohort or case-control studies. METHODS: A multicenter, randomized, controlled trial was conducted in 10 University Hospitals (NCT02803814, ClinicalTrials.gov). Eligible patients were those who presented with pancreatic head adenocarcinoma and periampullary tumors (ampulloma, distal cholangiocarcinoma, duodenal adenocarcinoma). Assignment to each group (ST-PD or AFA-PD) was randomized by blocks and stratified by centers. The primary end-point was the rate of tumor-free resection margins (R0); secondary end-points were postoperative complications and mortality. RESULTS: One hundred seventy-nine patients were assessed for eligibility and 176 randomized. After exclusions, the final analysis included 75 ST-PD and 78 AFA-PD. R0 resection rates were 77.3% (95% CI: 68.4-87.4) with ST-PD and 67.9% (95% CI: 58.3-79.1) with AFA-PD, P=0.194. There were no significant differences in postoperative complication rates, overall 73.3% versus 67.9%, and perioperative mortality 4% versus 6.4%. CONCLUSIONS: Despite theoretical oncological advantages associated with AFA-PD and evidence coming from low-level studies, this multicenter, randomized, controlled trial has found no difference neither in R0 resection rates nor in postoperative complications in patients undergoing ST-PD versus AFA-PD for pancreatic head adenocarcinoma and other periampullary tumors.


Asunto(s)
Adenocarcinoma/patología , Adenocarcinoma/cirugía , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/cirugía , Pancreaticoduodenectomía/métodos , Adenocarcinoma/mortalidad , Adulto , Anciano , Arterias/cirugía , Supervivencia sin Enfermedad , Femenino , Hospitales Universitarios , Humanos , Masculino , Márgenes de Escisión , Persona de Mediana Edad , Invasividad Neoplásica/patología , Estadificación de Neoplasias , Neoplasias Pancreáticas/mortalidad , Pancreaticoduodenectomía/efectos adversos , Pronóstico , Medición de Riesgo , Estadísticas no Paramétricas , Análisis de Supervivencia , Resultado del Tratamiento
10.
Blood ; 130(13): 1535-1542, 2017 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-28801449

RESUMEN

Previous Fanconi anemia (FA) gene therapy studies have failed to demonstrate engraftment of gene-corrected hematopoietic stem and progenitor cells (HSPCs) from FA patients, either after autologous transplantation or infusion into immunodeficient mice. In this study, we demonstrate that a validated short transduction protocol of G-CSF plus plerixafor-mobilized CD34+ cells from FA-A patients with a therapeutic FANCA-lentiviral vector corrects the phenotype of in vitro cultured hematopoietic progenitor cells. Transplantation of transduced FA CD34+ cells into immunodeficient mice resulted in reproducible engraftment of myeloid, lymphoid, and CD34+ cells. Importantly, a marked increase in the proportion of phenotypically corrected, patient-derived hematopoietic cells was observed after transplantation with respect to the infused CD34+ graft, indicating the proliferative advantage of corrected FA-A hematopoietic repopulating cells. Our data demonstrate for the first time that optimized protocols of hematopoietic stem cell collection from FA patients, followed by the short and clinically validated transduction of these cells with a therapeutic lentiviral vector, results in the generation of phenotypically corrected HSPCs capable of repopulating and developing proliferation advantage in immunodeficient mice. Our results suggest that clinical approaches for FA gene therapy similar to those used in this study will facilitate hematopoietic repopulation in FA patients with gene corrected HSPCs, opening new prospects for gene therapy of FA patients.


Asunto(s)
Proteína del Grupo de Complementación C de la Anemia de Fanconi/genética , Anemia de Fanconi/terapia , Terapia Genética/métodos , Vectores Genéticos , Trasplante de Células Madre Hematopoyéticas/métodos , Transducción Genética/métodos , Animales , Antígenos CD34/inmunología , Niño , Preescolar , Anemia de Fanconi/patología , Supervivencia de Injerto , Movilización de Célula Madre Hematopoyética , Células Madre Hematopoyéticas/patología , Xenoinjertos , Humanos , Lentivirus/genética , Ratones
11.
Cytotherapy ; 2024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38944796
12.
Rev Esp Enferm Dig ; 110(10): 669-671, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29931993

RESUMEN

Several types of thoracic complications can be associated with severe acute pancreatitis. Some are very common, such as pleural effusion, whilst some others are very rare although life threatening, such as enzymatic mediastinitis (EM). Only a few cases of EM (when related to acute pancreatitis) have been described in the literature. Here we describe the case of a 76-year-old female who developed acute respiratory failure and atrial fibrillation during the postoperative period after an open pancreatic necrosectomy, due to an EM episode. The mediastinal collection was drained by thoracotomy, following an improvement of the patient's general condition. This is the first case of EM following surgical management for acute necrotizing pancreatitis. EM is a rare but life threatening complication that usually requires surgery.


Asunto(s)
Mediastinitis , Pancreatitis Aguda Necrotizante/cirugía , Complicaciones Posoperatorias , Anciano , Femenino , Humanos , Mediastinitis/diagnóstico , Mediastinitis/enzimología , Mediastinitis/terapia , Páncreas/enzimología , Complicaciones Posoperatorias/diagnóstico , Complicaciones Posoperatorias/terapia
13.
Am J Hum Genet ; 92(5): 800-6, 2013 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-23623386

RESUMEN

Fanconi anemia (FA) is a rare genomic instability disorder characterized by progressive bone marrow failure and predisposition to cancer. FA-associated gene products are involved in the repair of DNA interstrand crosslinks (ICLs). Fifteen FA-associated genes have been identified, but the genetic basis in some individuals still remains unresolved. Here, we used whole-exome and Sanger sequencing on DNA of unclassified FA individuals and discovered biallelic germline mutations in ERCC4 (XPF), a structure-specific nuclease-encoding gene previously connected to xeroderma pigmentosum and segmental XFE progeroid syndrome. Genetic reversion and wild-type ERCC4 cDNA complemented the phenotype of the FA cell lines, providing genetic evidence that mutations in ERCC4 cause this FA subtype. Further biochemical and functional analysis demonstrated that the identified FA-causing ERCC4 mutations strongly disrupt the function of XPF in DNA ICL repair without severely compromising nucleotide excision repair. Our data show that depending on the type of ERCC4 mutation and the resulting balance between both DNA repair activities, individuals present with one of the three clinically distinct disorders, highlighting the multifunctional nature of the XPF endonuclease in genome stability and human disease.


Asunto(s)
Proteínas de Unión al ADN/genética , Desoxirribonucleasas/genética , Anemia de Fanconi/genética , Predisposición Genética a la Enfermedad/genética , Fenotipo , Apoptosis/genética , Apoptosis/efectos de la radiación , Secuencia de Bases , Exoma/genética , Anemia de Fanconi/patología , Mutación de Línea Germinal/genética , Humanos , Immunoblotting , Inmunoprecipitación , Datos de Secuencia Molecular , Análisis de Secuencia de ADN , Rayos Ultravioleta
14.
J Pathol ; 236(2): 219-28, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25664618

RESUMEN

BCR-JAK2 is an infrequent gene fusion found in chronic/acute, myeloid/lymphoid Philadelphia chromosome-negative leukaemia. In this study, we demonstrated that in vivo expression of BCR-JAK2 in mice induces neoplasia, with fatal consequences. Transplantation of BCR-JAK2 bone marrow progenitors promoted splenomegaly, with megakaryocyte infiltration and elevated leukocytosis of myeloid origin. Analysis of peripheral blood revealed the presence of immature myeloid cells, platelet aggregates and ineffective erythropoiesis. A possible molecular mechanism for these observations involved inhibition of apoptosis by deregulated expression of the anti-apoptotic mediator Bcl-xL and the serine/threonine kinase Pim1. Together, these data provide a suitable in vivo molecular mechanism for leukaemia induction by BCR-JAK2 that validates the use of this model as a relevant preclinical tool for the design of new targeted therapies in Philadelphia chromosome-negative leukaemia involving BCR-JAK2-driven activation of the JAK2 pathway.


Asunto(s)
Janus Quinasa 2/fisiología , Leucemia Mieloide Crónica Atípica BCR-ABL Negativa/genética , Proteínas Proto-Oncogénicas c-bcr/fisiología , Animales , Femenino , Reordenamiento Génico , Trasplante de Células Madre Hematopoyéticas/métodos , Células Madre Hematopoyéticas/fisiología , Janus Quinasa 2/genética , Leucemia Mieloide Crónica Atípica BCR-ABL Negativa/mortalidad , Leucocitosis/etiología , Masculino , Ratones Endogámicos BALB C , Trasplante de Neoplasias , Proteínas Proto-Oncogénicas c-bcr/genética , Retroviridae , Factor de Transcripción STAT5/metabolismo , Esplenomegalia/etiología , Transducción Genética/métodos , Transgenes
15.
Nature ; 460(7251): 53-9, 2009 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-19483674

RESUMEN

The generation of induced pluripotent stem (iPS) cells has enabled the derivation of patient-specific pluripotent cells and provided valuable experimental platforms to model human disease. Patient-specific iPS cells are also thought to hold great therapeutic potential, although direct evidence for this is still lacking. Here we show that, on correction of the genetic defect, somatic cells from Fanconi anaemia patients can be reprogrammed to pluripotency to generate patient-specific iPS cells. These cell lines appear indistinguishable from human embryonic stem cells and iPS cells from healthy individuals. Most importantly, we show that corrected Fanconi-anaemia-specific iPS cells can give rise to haematopoietic progenitors of the myeloid and erythroid lineages that are phenotypically normal, that is, disease-free. These data offer proof-of-concept that iPS cell technology can be used for the generation of disease-corrected, patient-specific cells with potential value for cell therapy applications.


Asunto(s)
Anemia de Fanconi/patología , Anemia de Fanconi/terapia , Células Madre Hematopoyéticas/citología , Células Madre Pluripotentes/citología , Línea Celular , Reprogramación Celular , Salud , Células Madre Hematopoyéticas/metabolismo , Humanos , Células Madre Pluripotentes/metabolismo
16.
Blood ; 119(5): 1139-50, 2012 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-22117040

RESUMEN

In vivo lentiviral vector (LV)-mediated gene delivery would represent a great step forward in the field of gene therapy. Therefore, we have engineered a novel LV displaying SCF and a mutant cat endogenous retroviral glycoprotein, RDTR. These RDTR/SCF-LVs outperformed RDTR-LVs for transduction of human CD34(+) cells (hCD34(+)). For in vivo gene therapy, these novel RDTR/SCF-displaying LVs can distinguish between the target hCD34(+) cells of interest and nontarget cells. Indeed, they selectively targeted transduction to 30%-40% of the hCD34(+) cells in cord blood mononuclear cells and in the unfractionated BM of healthy and Fanconi anemia donors, resulting in the correction of CD34(+) cells in the patients. Moreover, RDTR/SCF-LVs targeted transduction to CD34(+) cells with 95-fold selectivity compared with T cells in total cord blood. Remarkably, in vivo injection of the RDTR/SCF-LVs into the BM cavity of humanized mice resulted in the highly selective transduction of candidate hCD34(+)Lin(-) HSCs. In conclusion, this new LV will facilitate HSC-based gene therapy by directly targeting these primitive cells in BM aspirates or total cord blood. Most importantly, in the future, RDTR/SCF-LVs might completely obviate ex vivo handling and simplify gene therapy for many hematopoietic defects because of their applicability to direct in vivo inoculation.


Asunto(s)
Médula Ósea/metabolismo , Terapia Genética/métodos , Vectores Genéticos/fisiología , Células Madre Hematopoyéticas/metabolismo , Hemoglobinuria Paroxística/terapia , Lentivirus/genética , Anemia Aplásica , Animales , Animales Recién Nacidos , Médula Ósea/patología , Enfermedades de la Médula Ósea , Trastornos de Fallo de la Médula Ósea , Células Cultivadas , Proteínas de Unión al ADN/genética , Marcación de Gen/métodos , Técnicas de Transferencia de Gen , Vectores Genéticos/genética , Hemoglobinuria Paroxística/genética , Hemoglobinuria Paroxística/patología , Humanos , Cadenas gamma de Inmunoglobulina/genética , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Transducción Genética
17.
Blood ; 119(13): 3042-9, 2012 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-22310912

RESUMEN

Fanconi anemia (FA) is an inherited genetic disorder associated with BM failure and cancer predisposition. In the present study, we sought to elucidate the role of microRNAs (miRNAs) in the hematopoietic defects observed in FA patients. Initial studies showed that 3 miRNAs, hsa-miR-133a, hsa-miR-135b, and hsa-miR-181c, were significantly down-regulated in lymphoblastoid cell lines and fresh peripheral blood cells from FA patients. In vitro studies with cells expressing the luciferase reporter fused to the TNFα 3'-untranslated region confirmed in silico predictions suggesting an interaction between hsa-miR-181c and TNFα mRNA. These observations were consistent with the down-regulated expression of TNFα mediated by hsa-miR-181c in cells from healthy donors and cells from FA patients. Because of the relevance of TNFα in the hematopoietic defects of FA patients, in the present study, we transfected BM cells from FA patients with hsa-miR-181c to evaluate the impact of this miRNA on their clonogenic potential. hsa-miR-181c markedly increased the number and size of the myeloid and erythroid colonies generated by BM cells from FA patients. Our results offer new clues toward understanding the biologic basis of BM failure in FA patients and open new possibilities for the treatment of the hematologic dysfunction in FA patients based on miRNA regulation.


Asunto(s)
Proliferación Celular , Anemia de Fanconi/genética , Células Madre Hematopoyéticas/fisiología , MicroARNs/genética , Factor de Necrosis Tumoral alfa/farmacología , Recuento de Células Sanguíneas , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/fisiología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Regulación hacia Abajo/genética , Anemia de Fanconi/metabolismo , Expresión Génica/efectos de los fármacos , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Humanos , Masculino , MicroARNs/metabolismo , Cultivo Primario de Células , Transfección
18.
Nat Genet ; 37(9): 931-3, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16116424

RESUMEN

Seven Fanconi anemia-associated proteins (FANCA, FANCB, FANCC, FANCE, FANCF, FANCG and FANCL) form a nuclear Fanconi anemia core complex that activates the monoubiquitination of FANCD2, targeting FANCD2 to BRCA1-containing nuclear foci. Cells from individuals with Fanconi anemia of complementation groups D1 and J (FA-D1 and FA-J) have normal FANCD2 ubiquitination. Using genetic mapping, mutation identification and western-blot data, we identify the defective protein in FA-J cells as BRIP1 (also called BACH1), a DNA helicase that is a binding partner of the breast cancer tumor suppressor BRCA1.


Asunto(s)
Cromosomas Humanos Par 17 , Proteínas de Unión al ADN/genética , Anemia de Fanconi/genética , Mutación/genética , Polimorfismo de Nucleótido Simple , ARN Helicasas/genética , Ubiquitina/metabolismo , Western Blotting , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Núcleo Celular , Proteínas de Unión al ADN/metabolismo , Proteína del Grupo de Complementación C de la Anemia de Fanconi , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi , Proteínas del Grupo de Complementación de la Anemia de Fanconi , Femenino , Humanos , Masculino , Repeticiones de Microsatélite , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Linaje , ARN Helicasas/metabolismo
19.
JCI Insight ; 9(10)2024 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-38775150

RESUMEN

This study lays the groundwork for future lentivirus-mediated gene therapy in patients with Diamond Blackfan anemia (DBA) caused by mutations in ribosomal protein S19 (RPS19), showing evidence of a new safe and effective therapy. The data show that, unlike patients with Fanconi anemia (FA), the hematopoietic stem cell (HSC) reservoir of patients with DBA was not significantly reduced, suggesting that collection of these cells should not constitute a remarkable restriction for DBA gene therapy. Subsequently, 2 clinically applicable lentiviral vectors were developed. In the former lentiviral vector, PGK.CoRPS19 LV, a codon-optimized version of RPS19 was driven by the phosphoglycerate kinase promoter (PGK) already used in different gene therapy trials, including FA gene therapy. In the latter one, EF1α.CoRPS19 LV, RPS19 expression was driven by the elongation factor alpha short promoter, EF1α(s). Preclinical experiments showed that transduction of DBA patient CD34+ cells with the PGK.CoRPS19 LV restored erythroid differentiation, and demonstrated the long-term repopulating properties of corrected DBA CD34+ cells, providing evidence of improved erythroid maturation. Concomitantly, long-term restoration of ribosomal biogenesis was verified using a potentially novel method applicable to patients' blood cells, based on ribosomal RNA methylation analyses. Finally, in vivo safety studies and proviral insertion site analyses showed that lentivirus-mediated gene therapy was nontoxic.


Asunto(s)
Anemia de Diamond-Blackfan , Terapia Genética , Vectores Genéticos , Células Madre Hematopoyéticas , Lentivirus , Proteínas Ribosómicas , Anemia de Diamond-Blackfan/terapia , Anemia de Diamond-Blackfan/genética , Humanos , Terapia Genética/métodos , Lentivirus/genética , Proteínas Ribosómicas/genética , Vectores Genéticos/genética , Células Madre Hematopoyéticas/metabolismo , Animales , Ratones , Masculino , Femenino , Ribosomas/metabolismo , Ribosomas/genética , Regiones Promotoras Genéticas , Mutación , Trasplante de Células Madre Hematopoyéticas/métodos
20.
Sci Transl Med ; 16(734): eadg7962, 2024 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-38354229

RESUMEN

Multiple myeloma is the second most common hematological malignancy in adults and remains an incurable disease. B cell maturation antigen (BCMA)-directed immunotherapy, including T cells bearing chimeric antigen receptors (CARs) and systemically injected bispecific T cell engagers (TCEs), has shown remarkable clinical activity, and several products have received market approval. However, despite promising results, most patients eventually become refractory and relapse, highlighting the need for alternative strategies. Engineered T cells secreting TCE antibodies (STAb) represent a promising strategy that combines the advantages of adoptive cell therapies and bispecific antibodies. Here, we undertook a comprehensive preclinical study comparing the therapeutic potential of T cells either expressing second-generation anti-BCMA CARs (CAR-T) or secreting BCMAxCD3 TCEs (STAb-T) in a T cell-limiting experimental setting mimicking the conditions found in patients with relapsed/refractory multiple myeloma. STAb-T cells recruited T cell activity at extremely low effector-to-target ratios and were resistant to inhibition mediated by soluble BCMA released from the cell surface, resulting in enhanced cytotoxic responses and prevention of immune escape of multiple myeloma cells in vitro. These advantages led to robust expansion and persistence of STAb-T cells in vivo, generating long-lived memory BCMA-specific responses that could control multiple myeloma progression in xenograft models, outperforming traditional CAR-T cells. These promising preclinical results encourage clinical testing of the BCMA-STAb-T cell approach in relapsed/refractory multiple myeloma.


Asunto(s)
Mieloma Múltiple , Receptores Quiméricos de Antígenos , Adulto , Humanos , Mieloma Múltiple/patología , Linfocitos T , Inmunoterapia Adoptiva/métodos , Antígeno de Maduración de Linfocitos B , Memoria Inmunológica , Recurrencia Local de Neoplasia/metabolismo , Receptores Quiméricos de Antígenos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA