Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
J Pediatr Hematol Oncol ; 46(6): e457-e462, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-38934620

RESUMEN

The clinical course for Hereditary Spherocytosis (HS) patients is highly varied, even within families with identical driving mutations. Here, we describe four siblings with HS attributed to an unreported SPTB mutation. All patients displayed an increased fraction of mitochondria-positive erythrocytes. This was associated with increased reactive oxygen species (ROS) generation and alteration to alterations to bioactive membrane lipids associated with oxidant stress. Given the early promise for mitophagy-inducing agents in sickle cell disease and ready availability of antioxidants, this concept warrants continued exploration as a disease-modifying factor and a potential target for therapy.


Asunto(s)
Lípidos de la Membrana , Mitocondrias , Estrés Oxidativo , Esferocitosis Hereditaria , Humanos , Esferocitosis Hereditaria/metabolismo , Esferocitosis Hereditaria/genética , Esferocitosis Hereditaria/patología , Masculino , Mitocondrias/metabolismo , Femenino , Lípidos de la Membrana/metabolismo , Niño , Preescolar , Especies Reactivas de Oxígeno/metabolismo , Mutación , Adolescente
3.
Am J Physiol Regul Integr Comp Physiol ; 315(4): R840-R847, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30067082

RESUMEN

Sickle cell disease (SCD) is caused by a mutation of the ß-globin gene (Ingram VM. Nature 180: 326-328, 1957), which triggers the polymerization of deoxygenated sickle hemoglobin (HbS). Approximately 100,000 SCD patients in the United States and millions worldwide (Piel FB, et al. PLoS Med 10: e1001484, 2013) suffer from chronic hemolytic anemia, painful crises, multisystem organ damage, and reduced life expectancy (Rees DC, et al. Lancet 376: 2018-2031, 2010; Serjeant GR. Cold Spring Harb Perspect Med 3: a011783, 2013). Hematopoietic stem cell transplantation can be curative, but the majority of patients do not have a suitable donor (Talano JA, Cairo MS. Eur J Haematol 94: 391-399, 2015). Advanced gene-editing technologies also offer the possibility of a cure (Goodman MA, Malik P. Ther Adv Hematol 7: 302-315, 2016; Lettre G, Bauer DE. Lancet 387: 2554-2564, 2016), but the likelihood that these strategies can be mobilized to treat the large numbers of patients residing in developing countries is remote. A pharmacological treatment to increase fetal hemoglobin (HbF) as a therapy for SCD has been a long-sought goal, because increased levels of HbF (α2γ2) inhibit the polymerization of HbS (Poillin WN, et al. Proc Natl Acad Sci USA 90: 5039-5043, 1993; Sunshine HR, et al. J Mol Biol 133: 435-467, 1979) and are associated with reduced symptoms and increased lifespan of SCD patients (Platt OS, et al. N Engl J Med 330: 1639-1644, 1994; Platt OS, et al. N Engl J Med 325: 11-16, 1991). Only two drugs, hydroxyurea and l-glutamine, are approved by the US Food and Drug Administration for treatment of SCD. Hydroxyurea is ineffective at HbF induction in ~50% of patients (Charache S, et al. N Engl J Med 332: 1317-1322, 1995). While polymerization of HbS has been traditionally considered the driving force in the hemolysis of SCD, the excessive reactive oxygen species generated from red blood cells, with further amplification by intravascular hemolysis, also are a major contributor to SCD pathology. This review highlights a new class of drugs, lysine-specific demethylase (LSD1) inhibitors, that induce HbF and reduce reactive oxygen species.


Asunto(s)
Anemia de Células Falciformes/tratamiento farmacológico , Inhibidores Enzimáticos/farmacología , Eritrocitos/efectos de los fármacos , Histona Demetilasas/antagonistas & inhibidores , Anemia de Células Falciformes/sangre , Anemia de Células Falciformes/enzimología , Animales , Biomarcadores/sangre , Modelos Animales de Enfermedad , Diseño de Fármacos , Eritrocitos/enzimología , Hemoglobina Fetal/metabolismo , Histona Demetilasas/metabolismo , Humanos , Ratones , Terapia Molecular Dirigida , Papio , Especies Reactivas de Oxígeno/sangre , Medición de Riesgo
4.
Epigenetics ; 10(5): 397-407, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25932923

RESUMEN

The mechanism responsible for developmental stage-specific regulation of γ-globin gene expression involves DNA methylation. Previous results have shown that the γ-globin promoter is nearly fully demethylated during fetal liver erythroid differentiation and partially demethylated during adult bone marrow erythroid differentiation. The hypothesis that 5-hydroxymethylcytosine (5 hmC), a known intermediate in DNA demethylation pathways, is involved in demethylation of the γ-globin gene promoter during erythroid differentiation was investigated by analyzing levels of 5-methylcytosine (5 mC) and 5 hmC at a CCGG site within the 5' γ-globin gene promoter region in FACS-purified cells from baboon bone marrow and fetal liver enriched for different stages of erythroid differentiation. Our results show that 5 mC and 5 hmC levels at the γ-globin promoter are dynamically modulated during erythroid differentiation with peak levels of 5 hmC preceding and/or coinciding with demethylation. The Tet2 and Tet3 dioxygenases that catalyze formation of 5 hmC are expressed during early stages of erythroid differentiation and Tet3 expression increases as differentiation proceeds. In baboon CD34+ bone marrow-derived erythroid progenitor cell cultures, γ-globin expression was positively correlated with 5 hmC and negatively correlated with 5 mC at the γ-globin promoter. Supplementation of culture media with Vitamin C, a cofactor of the Tet dioxygenases, reduced γ-globin promoter DNA methylation and increased γ-globin expression when added alone and in an additive manner in combination with either DNA methyltransferase or LSD1 inhibitors. These results strongly support the hypothesis that the Tet-mediated 5 hmC pathway is involved in developmental stage-specific regulation of γ-globin expression by mediating demethylation of the γ-globin promoter.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Citosina/análogos & derivados , Metilación de ADN/efectos de los fármacos , Células Eritroides/citología , Regiones Promotoras Genéticas/efectos de los fármacos , gamma-Globinas/metabolismo , 5-Metilcitosina/metabolismo , Animales , Animales Recién Nacidos , Antineoplásicos/farmacología , Ácido Ascórbico/farmacología , Azacitidina/análogos & derivados , Azacitidina/farmacología , Células de la Médula Ósea , Células Cultivadas , Citosina/metabolismo , Citosina/farmacología , Decitabina , Dioxigenasas/metabolismo , Células Eritroides/efectos de los fármacos , Células Eritroides/metabolismo , Histona Demetilasas/metabolismo , Humanos , Hidroxiurea/farmacología , Hígado/citología , Hígado/efectos de los fármacos , Papio anubis , Tranilcipromina/farmacología
5.
Exp Hematol ; 43(7): 546-53.e1-3, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25931013

RESUMEN

Increased levels of fetal hemoglobin are associated with decreased symptoms and increased lifespan in patients with sickle cell disease (SCD). Hydroxyurea, the only drug currently approved for SCD, is not effective in a large fraction of patients, and therefore, new agents are urgently needed. Recently it was found that lysine demethylase 1, an enzyme that removes monomethyl and dimethyl residues from the lysine 4 residue of histone H3, is a repressor of γ-globin gene expression. In this article, we have compared the ability of tranylcypromine (TCP) and a more potent TCP derivative, RN-1, to increase γ-globin expression in cultured baboon erythroid progenitor cells and in the SCD mouse model. The results indicate that the ability of RN-1 to induce F cells and γ-globin mRNA in SCD mice is similar to that of decitabine, the most powerful fetal hemoglobin-inducing drug known, and greater than that of either TCP or hydroxyurea. We conclude that RN-1 and other lysine demethylase 1 inhibitors may be promising new γ-globin-inducing agents for the treatment of SCD that warrant further studies in other preclinical models, such as nonhuman primates.


Asunto(s)
Anemia de Células Falciformes/genética , Hemoglobina Fetal/biosíntesis , Histona Demetilasas/antagonistas & inhibidores , Reticulocitos/efectos de los fármacos , Tranilcipromina/farmacología , gamma-Globinas/biosíntesis , Anemia de Células Falciformes/tratamiento farmacológico , Anemia de Células Falciformes/enzimología , Animales , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Inhibidores Enzimáticos/farmacología , Células Precursoras Eritroides/efectos de los fármacos , Células Precursoras Eritroides/metabolismo , Eritropoyesis/efectos de los fármacos , Hemoglobina Fetal/genética , Regulación de la Expresión Génica/efectos de los fármacos , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Histonas/metabolismo , Humanos , Hidroxiurea/farmacología , Metilación , Ratones , Ratones Transgénicos , Papio , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Transgenes/efectos de los fármacos , Tranilcipromina/análogos & derivados , Tretinoina/farmacología , Células U937 , gamma-Globinas/genética
6.
Immunol Cell Biol ; 92(2): 116-23, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24217810

RESUMEN

Dendritic cell (DC)-based immunotherapy has recently demonstrated a great potential for clinical applications; however, additional progress in the methods of tumor-specific antigen delivery to DCs is necessary for the further development of anti-tumor vaccines. To this end, a capsid-optimized adeno-associated virus serotype 6 (AAV6-T492V+S663V) vector was developed by site-directed mutagenesis of surface-exposed serine (S) and threonine (T) residues, which have a critical role in intracellular trafficking of AAV vectors. This double-mutant AAV6 vector had ∼ 5-fold greater transduction efficiency in monocyte-derived DCs (moDCs) compared with wild-type (WT)-AAV6 vectors. The increase in the transduction efficiency correlated with the improved nuclear translocation of AAV6-T492V+S663V over that of the WT-AAV6 vector. Additional studies of the CD11c promoter identified critical regulatory elements that fit into the AAV expression cassette and drive EGFP expression in moDCs. Development of a chimeric promoter (chmCD11c) that contains functional modules of CD11c and a Simian virus (SV40) enhancer element dramatically increased the EGFP expression in moDCs. MoDCs transduced by the capsid-optimized AAV6 vector carrying human prostate-specific antigen (hPSA) driven by CBA (AAV6-T492V+S663V-CBA-hPSA) or chmCd11c (AAV6-T492V+S663V-chmCD11c-hPSA) generated specific T-cell clone proliferation and superior cytotoxic T lymphocytes (CTLs) with higher killing capability against human prostate adenocarcinoma cells, LNCaP, compared with WT-AAV6 induced CTLs. Taken together, these studies suggest that optimization of capsid and promoter components of AAV vectors can be a useful approach for efficient targeting of moDCs and may prove to be a promising tool for cancer immunotherapy.


Asunto(s)
Adenocarcinoma/terapia , Cápside/inmunología , Células Dendríticas/inmunología , Dependovirus , Vectores Genéticos , Inmunoterapia/métodos , Neoplasias de la Próstata/terapia , Transgenes , Adenocarcinoma/genética , Adenocarcinoma/inmunología , Línea Celular Tumoral , Humanos , Masculino , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/inmunología
7.
J Control Release ; 171(2): 184-92, 2013 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-23871960

RESUMEN

Selective drug delivery to hypoxic tumor niches remains a significant therapeutic challenge that calls for new conceptual approaches. Sickle red blood cells (SSRBCs) have shown an ability to target such hypoxic niches and induce tumoricidal effects when used together with exogenous pro-oxidants. Here we determine whether the delivery of a model therapeutic encapsulated in murine SSRBCs can be enhanced by ex vivo photosensitization under conditions that delay autohemolysis to a time that coincides with maximal localization of SSRBCs in a hypoxic tumor. Hyperspectral imaging of 4T1 carcinomas shows oxygen saturation levels <10% in a large fraction (commonly 50% or more) of the tumor. Using video microscopy of dorsal skin window chambers implanted with 4T1 tumors, we demonstrate that allogeneic SSRBCs, but not normal RBCs (nRBCs), selectively accumulate in hypoxic 4T1 tumors between 12 and 24h after systemic administration. We further show that ex vivo photo-oxidation can program SSRBCs to postpone hemolysis/release of a model therapeutic to a point that coincides with their maximum sequestration in hypoxic tumor microvessels. Under these conditions, drug-loaded photosensitized SSRBCs show a 3-4 fold greater drug delivery to tumors compared to non-photosensitized SSRBCs, drug-loaded photosensitized nRBCs, and free drug. These results demonstrate that photo-oxidized SSRBCs, but not photo-oxidized nRBCs, sequester and hemolyze in hypoxic tumors and release substantially more drug than photo-oxidized nRBCs and non-photo-oxidized SSRBCs. Photo-oxidation of drug-loaded SSRBCs thus appears to exploit the unique tumor targeting and carrier properties of SSRBCs to optimize drug delivery to hypoxic tumors. Such programmed and drug-loaded SSRBCs therefore represent a novel and useful tool for augmenting drug delivery to hypoxic solid tumors.


Asunto(s)
Anemia de Células Falciformes , Sistemas de Liberación de Medicamentos , Eritrocitos , Neoplasias/metabolismo , Animales , Línea Celular Tumoral , Eritrocitos/efectos de los fármacos , Eritrocitos/efectos de la radiación , Femenino , Colorantes Fluorescentes/administración & dosificación , Colorantes Fluorescentes/farmacocinética , Hemólisis , Humanos , Hipoxia , Luz , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Ratones Transgénicos , Microvasos , Neoplasias/irrigación sanguínea , Oxidación-Reducción , Fotólisis , Fármacos Fotosensibilizantes/farmacología , Protoporfirinas/farmacología , Bazo/metabolismo
8.
Cytotherapy ; 15(8): 986-98, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23830234

RESUMEN

BACKGROUND AIMS: Although recombinant adeno-associated virus serotype 2 (AAV2) vectors have gained attention because of their safety and efficacy in numerous phase I/II clinical trials, their transduction efficiency in hematopoietic stem cells (HSCs) has been reported to be low. Only a few additional AAV serotype vectors have been evaluated, and comparative analyses of their transduction efficiency in HSCs from different species have not been performed. METHODS: We evaluated the transduction efficiency of all available AAV serotype vectors (AAV1 through AAV10) in primary mouse, cynomolgus monkey and human HSCs. The transduction efficiency of the optimized AAV vectors was also evaluated in human HSCs in a murine xenograft model in vivo. RESULTS: We observed that although there are only six amino acid differences between AAV1 and AAV6, AAV1, but not AAV6, transduced mouse HSCs well, whereas AAV6, but not AAV1, transduced human HSCs well. None of the 10 serotypes transduced cynomolgus monkey HSCs in vitro. We also evaluated the transduction efficiency of AAV6 vectors containing mutations in surface-exposed tyrosine residues. We observed that tyrosine (Y) to phenylalanine (F) point mutations in residues 445, 705 and 731 led to a significant increase in transgene expression in human HSCs in vitro and in a mouse xenograft model in vivo. CONCLUSIONS: These studies suggest that the tyrosine-mutant AAV6 serotype vectors are the most promising vectors for transducing human HSCs and that it is possible to increase further the transduction efficiency of these vectors for their potential use in HSC-based gene therapy in humans.


Asunto(s)
Terapia Genética/métodos , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas , Transducción Genética/métodos , Animales , Antígenos CD34/metabolismo , Línea Celular , Dependovirus , Expresión Génica , Vectores Genéticos , Células HEK293 , Humanos , Células K562 , Macaca fascicularis , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones SCID
9.
PLoS One ; 8(3): e59142, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23527116

RESUMEN

The ubiquitin-proteasome pathway plays a critical role in the intracellular trafficking of AAV2 vectors, and phosphorylation of certain surface-exposed amino acid residues on the capsid provides the primary signal for ubiquitination. Removal of several critical tyrosine (Y) and serine (S) residues on the AAV2 capsid has been shown to significantly increase transduction efficiency compared with the wild-type (WT) vectors. In the present study, site-directed mutagenesis of each of the 17 surface-exposed threonine (T) residues was conducted, and the transduction efficiency of four of these mutants, T455V, T491V, T550V, and T659V, was observed to increase up to 4-fold in human HEK293 cells in vitro. The most critical Y, S, and T mutations were subsequently combined, and the quadruple-mutant (Y444+500+730F+T491V) AAV2 vector was identified as the most efficient. This vector increased the transduction efficiency ∼24-fold over the WT AAV2 vector, and ∼2-3-fold over the previously described triple-mutant (Y444+500+730F) vector in a murine hepatocyte cell line, H2.35, in vitro. Similar results were obtained in murine hepatocytes in vivo following tail vein injection of the Y444+500+730F+T491V scAAV2 vector, and whole-body bioluminescence imaging of C57BL/6 mice. The increase in the transduction efficiency of the Y-T quadruple-mutant over that of the Y triple-mutant correlated with an improved nuclear translocation of the vectors, which exceeded 90%. These observations suggest that further optimization of the AAV2 capsid by targeting amino acid residues involved in phosphorylation may not be possible. This study has thus led to the generation of a novel Y444+500+730F+T491V quadruple-mutant AAV2 vector with potential for use in liver-directed human gene therapy.


Asunto(s)
Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , Dependovirus/genética , Dependovirus/metabolismo , Vectores Genéticos/genética , Sustitución de Aminoácidos , Animales , Transporte Biológico , Línea Celular , Núcleo Celular/metabolismo , Expresión Génica , Hepatocitos/metabolismo , Hepatocitos/virología , Humanos , Masculino , Ratones , Mutagénesis Sitio-Dirigida , Treonina , Transducción Genética , Transgenes
10.
Vaccine ; 30(26): 3908-17, 2012 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-22497875

RESUMEN

Phosphorylation of surface-exposed tyrosine residues negatively impacts the transduction efficiency of recombinant AAV2 vectors. Pre-treatment of cells with specific cellular serine/threonine kinase inhibitors also significantly increased the transduction efficiency of AAV2 vectors. We reasoned that site-directed mutagenesis of surface-exposed serine residues might allow the vectors to evade phosphorylation and thus lead to higher transduction efficiency. Each of the 15 surface-exposed serine (S) residues was substituted with valine (V) residues, and the transduction efficiency of three of these mutants, S458V, S492V and S662V, was increased by up to ≈ 20-fold in different cell types. The S662V mutant was efficient in transducing human monocyte-derived dendritic cells (moDCs), a cell type not readily amenable to transduction by the conventional AAV vectors, and did not induce any phenotypic changes in these cells. Recombinant S662V-AAV2 vectors encoding a truncated human telomerase (hTERT) gene were generated and used to stimulate cytotoxic T cells (CTLs) against target cells. S662V-AAV2-hTERT vector-transduced DCs resulted in rapid, specific T-cell clone proliferation and generation of robust CTLs, which led to specific cell lysis of K562 cells. These studies suggest that high-efficiency transduction of moDCs by serine-modified AAV2 vectors is feasible, which supports the potential utility of these vectors for future human DCs vaccine studies.


Asunto(s)
Adenovirus Humanos/genética , Proteínas de la Cápside/genética , Células Dendríticas/virología , Vectores Genéticos , Transducción Genética , Sustitución de Aminoácidos , Células Cultivadas , Humanos , Mutagénesis Sitio-Dirigida , Proteínas Mutantes/genética , Serina/genética
11.
Hum Gene Ther ; 21(3): 271-83, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19788390

RESUMEN

Abstract Our studies have shown that coinjection of conventional single-stranded adeno-associated virus 2 (ssAAV2) vectors carrying the enhanced green fluorescent protein (EGFP) gene with self-complementary (sc) AAV2-T cell protein tyrosine phosphatase (TC-PTP) and scAAV2-protein phosphatase-5 (PP5) vectors resulted in an approximately 16-fold increase in EGFP expression in primary murine hepatocytes in vivo [Jayandharan, G.R., Zhong, L., Li, B., Kachniarz, B., and Srivastava, A. (2008). Gene Ther. 15, 1287-1293]. In the present studies, this strategy was further optimized to achieve transgene expression at reduced vector/helper virus doses. These included the use of scAAV helper viruses containing (1) hepatocyte-specific promoters, (2) tyrosine-mutant AAV2 capsids, and (3) additional AAV serotype vectors known to efficiently transduce hepatocytes. The hepatocyte-specific transthyretin (TTR) promoter was approximately 6- to 7-fold more efficient than the Rous sarcoma virus (RSV) promoter; tyrosine-mutant AAV2 capsids were approximately 6- to 11-fold more efficient than the wild-type AAV2 capsids; and the AAV8 serotype helper virus was approximately 16-fold more efficient than AAV2 serotype helper virus. With these modifications, the vector dose of the helper virus could be further reduced by approximately 50-fold. Last, coadministration of scAAV8-PP5 helper virus increased coagulation factor IX expression from an ssAAV2 vector by approximately 7- to 10-fold, thereby achieving therapeutic levels at lower vector doses. No adverse effect on hepatocytes was observed under any of these experimental conditions. The strategy presented here should be adaptable to any ssAAV transgene cassette and, specifically, liver-directed applications of ssAAV2 vectors containing larger genes that cannot be encapsidated in scAAV vectors.


Asunto(s)
Dependovirus/genética , Factor IX/genética , Vectores Genéticos , Virus Helper/genética , Hígado/metabolismo , Proteínas Nucleares/genética , Fosfoproteínas Fosfatasas/genética , Transducción Genética , Transgenes/fisiología , Animales , ADN de Cadena Simple , Técnicas de Transferencia de Gen , Proteínas Fluorescentes Verdes/genética , Hepatocitos/metabolismo , Hepatocitos/patología , Hepatocitos/virología , Humanos , Técnicas para Inmunoenzimas , Hígado/patología , Hígado/virología , Masculino , Ratones , Ratones Endogámicos C57BL , Prealbúmina/genética , Regiones Promotoras Genéticas , Proteína Tirosina Fosfatasa no Receptora Tipo 2/genética
12.
Semin Perinatol ; 33(1): 20-8, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19167578

RESUMEN

Congenital bone marrow failure syndromes (CBMFS) are extremely uncommon diseases that can present in the neonate. The objective of this article is to review the presentation, diagnosis, pathophysiology, and management of CBMFS in relation to neonatology. CBMFS should be considered when a single or multiple blood cell lineages are low secondary to failure of production. Diagnosis in the neonatal period requires a high index of suspicion. In this particular age group, CBMFS should be considered when the neonate has a family history of CBMFS, is small for gestational age, or has other physical abnormalities. History and physical examination can lead to the diagnosis. CBMFS are often associated with a predisposition to cancer later in life.


Asunto(s)
Anemia/congénito , Enfermedades de la Médula Ósea/congénito , Neutropenia/congénito , Trombocitopenia/congénito , Anemia/diagnóstico , Anemia/fisiopatología , Anemia/terapia , Enfermedades de la Médula Ósea/diagnóstico , Enfermedades de la Médula Ósea/fisiopatología , Enfermedades de la Médula Ósea/terapia , Humanos , Recién Nacido , Neutropenia/diagnóstico , Neutropenia/fisiopatología , Neutropenia/terapia , Trombocitopenia/diagnóstico , Trombocitopenia/fisiopatología , Trombocitopenia/terapia
13.
J Pediatr Hematol Oncol ; 30(9): 708-11, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18776767

RESUMEN

We report a child with thrombocytopenia since birth, circulating platelet agglutinates, and a tendency to bleed. A bone marrow aspirate revealed large platelet clumps within the bone marrow and megakaryocyte nuclei surrounded by halos of clumped platelets. Laboratory evaluation revealed type 2B von Willebrand disease. Gene sequencing revealed a G to C mutation at base 3923 of the VWF gene. This mutation was previously described in a family with circulating platelet clumps and abnormal megakaryopoiesis with release of clumped platelets in culture. This same mutation was previously described in a family with circulating platelet aggregates and abnormalities of platelet release from megakaryocytes in vitro. Presence of megakaryocytes with halos of clumped platelets in our patient suggests that platelet agglutinate occurs in the bone marrow in some type 2B von Willebrand disease patients.


Asunto(s)
Plaquetas/patología , Megacariocitos/patología , Enfermedades de von Willebrand/patología , Aglutinación , Examen de la Médula Ósea , Niño , Humanos , Masculino , Mutación , Trombocitopenia/complicaciones , Enfermedades de von Willebrand/diagnóstico , Factor de von Willebrand/genética
14.
Hum Gene Ther ; 19(4): 365-75, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18399730

RESUMEN

Therapeutic levels of expression of the beta-globin gene have been difficult to achieve with conventional retroviral vectors without the inclusion of DNase I-hypersensitive site (HS2, HS3, and HS4) enhancer elements. We generated recombinant adeno-associated viral (AAV) vectors carrying an antisickling human beta-globin gene under the control of either the beta-globin gene promoter/enhancer or the erythroid cell-specific human parvovirus B19 promoter at map unit 6 (B19p6) without any enhancer, and tested their efficacy in a human erythroid cell line (K-562) and in primary murine hematopoietic progenitor cells (c-kit(+)lin()). We report here that (1) self-complementary AAV serotype 2 (scAAV2)-beta-globin vectors containing only the HS2 enhancer are more efficient than single-stranded AAV (ssAAV2)-beta-globin vectors containing the HS2+HS3+HS4 enhancers; (2) scAAV2-beta-globin vectors recombine with scAAV2-HS2+HS3+HS4 vectors after dual-vector transduction, leading to transgene expression; (3) scAAV2-beta-globin as well as scAAV1-beta-globin vectors containing the B19p6 promoter without the HS2 enhancer element are more efficient than their counterparts containing the HS2 enhancer/beta-globin promoter; and (4) scAAV2-B19p6-beta-globin vectors in K-562 cells, and scAAV1-B19p6-beta-globin vectors in murine c-kit(+)lin() cells, yield efficient expression of the beta-globin protein. Thus, the combined use of scAAV vectors and the parvovirus B19 promoter may lead to expression of therapeutic levels the beta-globin gene in human erythroid cells, which has implications in the use of these vectors in gene therapy of beta-thalassemia and sickle cell disease.


Asunto(s)
Dependovirus/genética , Terapia Genética , Vectores Genéticos/genética , Globinas/genética , Transducción Genética , Transgenes/genética , Animales , Línea Celular , Elementos de Facilitación Genéticos/genética , Expresión Génica , Células Madre Hematopoyéticas/metabolismo , Humanos , Ratones , Parvovirus B19 Humano , Regiones Promotoras Genéticas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA