Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Am J Hum Genet ; 99(1): 115-24, 2016 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-27346687

RESUMEN

A substantial number of individuals with bone marrow failure (BMF) present with one or more extra-hematopoietic abnormality. This suggests a constitutional or inherited basis, and yet many of them do not fit the diagnostic criteria of the known BMF syndromes. Through exome sequencing, we have now identified a subgroup of these individuals, defined by germline biallelic mutations in DNAJC21 (DNAJ homolog subfamily C member 21). They present with global BMF, and one individual developed a hematological cancer (acute myeloid leukemia) in childhood. We show that the encoded protein associates with rRNA and plays a highly conserved role in the maturation of the 60S ribosomal subunit. Lymphoblastoid cells obtained from an affected individual exhibit increased sensitivity to the transcriptional inhibitor actinomycin D and reduced amounts of rRNA. Characterization of mutations revealed impairment in interactions with cofactors (PA2G4, HSPA8, and ZNF622) involved in 60S maturation. DNAJC21 deficiency resulted in cytoplasmic accumulation of the 60S nuclear export factor PA2G4, aberrant ribosome profiles, and increased cell death. Collectively, these findings demonstrate that mutations in DNAJC21 cause a cancer-prone BMF syndrome due to corruption of early nuclear rRNA biogenesis and late cytoplasmic maturation of the 60S subunit.


Asunto(s)
Anemia Aplásica/complicaciones , Anemia Aplásica/genética , Enfermedades de la Médula Ósea/complicaciones , Enfermedades de la Médula Ósea/genética , Proteínas del Choque Térmico HSP40/genética , Hemoglobinuria Paroxística/complicaciones , Hemoglobinuria Paroxística/genética , Mutación/genética , Neoplasias/complicaciones , Neoplasias/genética , Subunidades Ribosómicas Grandes de Eucariotas/metabolismo , Subunidades Ribosómicas Grandes de Eucariotas/patología , Secuencia de Aminoácidos , Trastornos de Fallo de la Médula Ósea , Proliferación Celular , Forma de la Célula , Niño , Preescolar , Femenino , Proteínas del Choque Térmico HSP40/química , Humanos , Leucemia Mieloide Aguda/complicaciones , Leucemia Mieloide Aguda/genética , Masculino , Unión Proteica , ARN Ribosómico/biosíntesis
2.
Br J Haematol ; 172(3): 360-70, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26577457

RESUMEN

Allogeneic haemopoietic stem cell transplantation offers a potentially curative treatment option for a wide range of life-threatening malignant and non-malignant disorders of the bone marrow and immune system in patients of all ages. With rapidly emerging advances in the use of alternative donors, such as mismatched unrelated, cord blood and haploidentical donors, it is now possible to find a potential donor for almost all patients in whom an allograft is indicated. Therefore, for any specific patient, the transplant physician may be faced with a myriad of potential choices, including decisions concerning which donor to prioritize where there is more than one, the optimal selection of specific umbilical cord blood units and which conditioning and graft-versus-host disease prophylactic schedule to use. Donor choice may be further complicated by other important factors, such as urgency of transplant, the presence of alloantibodies, the disease status (homozygosity or heterozygosity) of sibling donors affected by inherited disorders and the cytomegalovirus serostatus of patient and donor. We report UK consensus guidelines on the selection of umbilical cord blood units, the hierarchy of donor selection and the preferred conditioning regimens for umbilical cord blood transplantation, with a summary of rationale supporting these recommendations.


Asunto(s)
Trasplante de Células Madre de Sangre del Cordón Umbilical/normas , Selección de Donante , Acondicionamiento Pretrasplante/métodos , Algoritmos , Protocolos Clínicos , Trasplante de Células Madre de Sangre del Cordón Umbilical/métodos , Prueba de Histocompatibilidad/métodos , Humanos , Reino Unido
3.
Br J Haematol ; 171(4): 585-94, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26223288

RESUMEN

We explored the feasibility of unrelated donor haematopoietic stem cell transplant (HSCT) upfront without prior immunosuppressive therapy (IST) in paediatric idiopathic severe aplastic anaemia (SAA). This cohort was then compared to matched historical controls who had undergone first-line therapy with a matched sibling/family donor (MSD) HSCT (n = 87) or IST with horse antithymocyte globulin and ciclosporin (n = 58) or second-line therapy with unrelated donor HSCT post-failed IST (n = 24). The 2-year overall survival in the upfront cohort was 96 ± 4% compared to 91 ± 3% in the MSD controls (P = 0·30) and 94 ± 3% in the IST controls (P = 0·68) and 74 ± 9% in the unrelated donor HSCT post-IST failure controls (P = 0·02).The 2-year event-free survival in the upfront cohort was 92 ± 5% compared to 87 ± 4% in MSD controls (P = 0·37), 40 ± 7% in IST controls (P = 0·0001) and 74 ± 9% in the unrelated donor HSCT post-IST failure controls (n = 24) (P = 0·02). Outcomes for upfront-unrelated donor HSCT in paediatric idiopathic SAA were similar to MSD HSCT and superior to IST and unrelated donor HSCT post-IST failure. Front-line therapy with matched unrelated donor HSCT is a novel treatment approach and could be considered as first-line therapy in selected paediatric patients who lack a MSD.


Asunto(s)
Anemia Aplásica/terapia , Trasplante de Médula Ósea/estadística & datos numéricos , Trasplante de Células Madre de Sangre Periférica/estadística & datos numéricos , Infecciones por Adenoviridae/tratamiento farmacológico , Infecciones por Adenoviridae/epidemiología , Adolescente , Adulto , Anemia Aplásica/mortalidad , Suero Antilinfocítico , Transfusión Sanguínea/estadística & datos numéricos , Trasplante de Médula Ósea/efectos adversos , Estudios de Casos y Controles , Niño , Preescolar , Ciclosporina/uso terapéutico , Supervivencia sin Enfermedad , Femenino , Estudios de Seguimiento , Enfermedad Injerto contra Huésped/tratamiento farmacológico , Enfermedad Injerto contra Huésped/epidemiología , Enfermedad Injerto contra Huésped/prevención & control , Infecciones por Herpesviridae/tratamiento farmacológico , Infecciones por Herpesviridae/epidemiología , Histocompatibilidad , Humanos , Inmunosupresores/uso terapéutico , Lactante , Estimación de Kaplan-Meier , Tiempo de Internación/estadística & datos numéricos , Donadores Vivos , Masculino , Trasplante de Células Madre de Sangre Periférica/efectos adversos , Complicaciones Posoperatorias/tratamiento farmacológico , Complicaciones Posoperatorias/epidemiología , Disfunción Primaria del Injerto/epidemiología , Calidad de Vida , Estudios Retrospectivos , Hermanos , Tasa de Supervivencia , Linfocitos T , Resultado del Tratamiento , Activación Viral , Adulto Joven
4.
Mol Ther ; 21(4): 868-76, 2013 04.
Artículo en Inglés | MEDLINE | ID: mdl-23423338

RESUMEN

Hematopoietic stem cell gene therapy for neurological disorders relies on transmigration of donor-derived monocytes to the brain, where they can engraft as microglia and deliver therapeutic proteins. Many mouse studies use whole-body irradiation to investigate brain transmigration pathways, but chemotherapy is generally used clinically. The current evidence for transmigration to the brain after chemotherapy is conflicting. We compared hematopoietic donor cell brain engraftment after bone marrow (BM) transplants in busulfan- or irradiation-conditioned mice. Significantly more donor-derived microglial cells engrafted posttransplant in busulfan-conditioned brain compared with the irradiated, in both the short and long term. Although total Iba-1(+) microglial content was increased in irradiated brain in the short term, it was similar between groups over long-term engraftment. MCP-1, a key regulator of monocyte transmigration, showed long-term elevation in busulfan-conditioned brain, whereas irradiated brains showed long-term elevation of the proinflammatory chemokine interleukin 1α (IL-1α), with increased in situ proliferation of resident microglia, and significant increases in the relative number of amoeboid activated microglia in the brain. This has implications for the choice of conditioning regimen to promote hematopoietic cell brain engraftment and the relevance of irradiation in mouse models of transplantation.


Asunto(s)
Encéfalo/efectos de los fármacos , Encéfalo/efectos de la radiación , Busulfano/uso terapéutico , Células Madre Hematopoyéticas/citología , Animales , Trasplante de Médula Ósea , Encéfalo/citología , Encéfalo/metabolismo , Quimiocina CCL2/metabolismo , Interleucina-1/metabolismo , Ratones , Acondicionamiento Pretrasplante , Irradiación Corporal Total
5.
Biol Blood Marrow Transplant ; 19(11): 1608-14, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24029650

RESUMEN

Little information is currently available regarding the pharmacokinetics (PK) of busulfan in infants and small children to help guide decisions for safe and efficacious drug therapy. The objective of this study was to develop an algorithm for individualized dosing of i.v. busulfan in infants and children weighing ≤12 kg, that would achieve targeted exposure with the first dose of busulfan. Population PK modeling was conducted using intensive time-concentration data collected through the routine therapeutic drug monitoring of busulfan in 149 patients from 8 centers. Busulfan PK was well described by a 1-compartment base model with linear elimination. The important clinical covariates affecting busulfan PK were actual body weight and age. Based on our model, the predicted clearance of busulfan increases approximately 1.7-fold between 6 weeks to 2 years of life. For infants age <5 months, the model-predicted doses (mg/kg) required to achieve a therapeutic concentration at steady state of 600-900 ng/mL (area under the curve range, 900-1350 µM·min) were much lower compared with standard busulfan doses of 1.1 mg/kg. These results could help guide clinicians and inform better dosing decisions for busulfan in young infants and small children undergoing hematopoietic cell transplantation.


Asunto(s)
Antineoplásicos Alquilantes/farmacocinética , Busulfano/farmacocinética , Trasplante de Células Madre Hematopoyéticas/métodos , Trasplante Autólogo/métodos , Antineoplásicos Alquilantes/uso terapéutico , Busulfano/uso terapéutico , Preescolar , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Estudios Retrospectivos , Acondicionamiento Pretrasplante
7.
JIMD Rep ; 64(5): 346-352, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37701322

RESUMEN

Metachromatic leukodystrophy (MLD) results from ARSA gene mutations. Affected individuals meet early milestones before neurological deterioration and early death. Atidarsagene autotemcel (arsa-cel), an autologous haematopoietic stem cell gene therapy (HSC-GT) product, has demonstrated sustained clinical benefits in MLD. Arsa-cel was approved for NHS treatment in February 2022 for asymptomatic late infantile or early juvenile disease, or early symptomatic early juvenile MLD. We evaluate the impact of this approval in the largest real-world dataset of MLD HSC-GT. Hospital records were reviewed for all patients referred for NHS treatment following arsa-cel approval. Information was gathered about disease phenotype, presentation, eligibility, and affected siblings. In the year following NHS approval, 17 UK MLD patients were referred for treatment. Four patients met eligibility criteria and have been treated, including 1 infant who weighed 5 kg at leukapheresis. Eleven patients failed screening: 10 symptomatic patients with late infantile disease and 1 with early juvenile disease and cognitive decline. Two further patients with later onset subtypes did not meet the approval criteria. Three out of four treated patients were diagnosed by screening after MLD was diagnosed in a symptomatic older sibling. The success of HSC-GT for MLD has heralded a new era of hope for families affected by this devastating disease, yet currently, most patients are ineligible for treatment at diagnosis. The feasibility of apheresis in infants and the availability of a licenced, effective HSC-GT product highlights the urgent need for newborn screening to ensure that patients can be diagnosed and treated before symptom onset.

8.
Br J Haematol ; 157(3): 339-46, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22372373

RESUMEN

We retrospectively analysed the outcome of consecutive children with idiopathic severe aplastic anaemia in the United Kingdom who received immunosuppressive therapy (IST) or matched unrelated donor (MUD) haematopoietic stem cell transplantation (HSCT). The 6-month cumulative response rate following rabbit antithymocyte globulin (ATG)/ciclosporin (IST) was 32·5% (95% CI 19·3-46·6) (n = 43). The 5-year estimated failure-free survival (FFS) following IST was 13·3% (95% confidence interval [CI] 4·0-27·8). In contrast, in 44 successive children who received a 10-antigen (HLA-A, -B, -C, -DRB1, -DQB1) MUD HSCT there was an excellent estimated 5-year FFS of 95·01% (95% CI 81·38-98·74). Forty of these children had failed IST previously. HSCT conditioning was a fludarabine, cyclophosphamide and alemtuzumab (FCC) regimen and did not include radiotherapy. There were no cases of graft failure. Median donor chimerism was 100% (range 88-100%). A conditioning regimen, such as FCC that avoids total body irradiation is ideally suited in children. Our data suggest that MUD HSCT following IST failure offers an excellent outcome and furthermore, if a suitable MUD can be found quickly, MUD HSCT may be a reasonable alternative to IST.


Asunto(s)
Anemia Aplásica/terapia , Trasplante de Células Madre Hematopoyéticas/métodos , Donante no Emparentado , Adolescente , Suero Antilinfocítico/uso terapéutico , Niño , Preescolar , Ciclosporina/uso terapéutico , Femenino , Enfermedad Injerto contra Huésped/etiología , Humanos , Inmunosupresores/uso terapéutico , Lactante , Estimación de Kaplan-Meier , Masculino , Infecciones Oportunistas/etiología , Recurrencia , Estudios Retrospectivos , Quimera por Trasplante , Acondicionamiento Pretrasplante/métodos , Insuficiencia del Tratamiento , Resultado del Tratamiento
9.
Front Cell Dev Biol ; 10: 836594, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35465327

RESUMEN

Umbilical Cord blood is an intuitively attractive stem cell source, but its use has declined since it is associated with an increased procedure-related morbidity and transplant related mortality. Some of this reflects that cord blood transplants are more often HLA-mismatched compared to other unrelated donor transplants. The ability to transplant in such a setting, indeed without high rates of chronic Graft versus Host Disease (GVHD), constitutes an advantage compared to other unrelated donor cell sources and there are other advantages specifically associated with cord blood as a donor cell source. These advantages must be weighed against its disadvantage, and we have utilised cord blood preferentially as a donor cell source in certain clinical situations in paediatric medicine. In non-malignant diseases, outcomes in metabolic disease are critically dependent on age at transplant and the enzyme delivered by that transplant, and in cord blood transplantation then the time to transplant can be minimised and the engrafted recipients have higher chimerism that delivers higher enzyme levels. In malignant diseases, studies have described reduced relapse rate and better GVHD-free survival, and so we have prioritised cord as a donor cell source where the risk of relapse is highest, and the effects of higher transplant related mortality is most clearly offset by the reduced relapse rates.

10.
Br J Haematol ; 152(3): 249-60, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21133886

RESUMEN

There are a number of options for salvage treatment in children and adolescents with relapsed and refractory classical Hodgkin Lymphoma. These include salvage with standard dose chemotherapy, high dose chemotherapy with autologous stem cell transplant, allogeneic stem cell transplant or other novel approach. Radiotherapy has an important role in the salvage of some patients as part of a combined modality approach. This review outlines these salvage approaches and discusses whether the evidence from paediatric studies justifies a risk-adapted approach to salvage for individual patients or whether all patients should receive consolidation with high dose chemotherapy and autologous stem cell transplantation, which is often described as standard salvage management in adults. The important prognostic factors and how these may be used to allocate patients to standard versus high dose chemotherapy regimens are discussed. The role of allogeneic transplantation, novel agents and late effects will also be discussed.


Asunto(s)
Enfermedad de Hodgkin/terapia , Terapia Recuperativa/métodos , Adolescente , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Niño , Terapia Combinada , Esquema de Medicación , Femenino , Trasplante de Células Madre Hematopoyéticas/métodos , Humanos , Infertilidad/prevención & control , Masculino , Pronóstico , Recurrencia
11.
J Inherit Metab Dis ; 34(2): 499-508, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21170681

RESUMEN

Early detection of mucopolysaccharidosis (MPS) is an important factor in treatment success; therefore, good disease biomarkers are vital. We evaluate heparin cofactor II-thrombin complex (HCII-T) as a biomarker in serum and dried blood spots (DBS) of MPS patients. Serum HCII-T and urine dermatan sulphate:chondroitin sulphate (DS:CS) ratio are also compared longitudinally against clinical outcomes in MPSI, II and VI patients following treatment. Samples were collected from MPS patients at the Royal Manchester Children's Hospital. DS:CS ratio was obtained by measuring the area density of spots from 2D electrophoresis of urinary glycosaminoglycans. Serum and DBS HCII-T was measured by sandwich ELISA. Serum HCII-T is elevated approximately 25-fold in MPS diseases that store DS, clearly distinguishing untreated MPSI, II and VI patients from unaffected age-matched controls. Serum HCII-T is also elevated in MPSIII, which leads to storage of heparan sulphate, with an increase of approximately 4-fold over unaffected age-matched controls. Urine DS:CS ratio and serum HCII-T decrease in response to treatment of MPSI, II and VI patients. HCII-T appears to respond rapidly to perturbations in treatment, whilst DS:CS ratio responds more slowly. HCII-T is a suitable biomarker for MPSI, II and VI, and it may also be informative for MPS diseases storing HS alone, such as MPSIII, although the elevation observed is smaller. In treated MPS patients, HCII-T and DS:CS ratio appear to measure short-term and long-term treatment outcomes, respectively. The potential value of HCII-T measurement in DBS for newborn screening of MPS diseases warrants further investigation.


Asunto(s)
Sulfatos de Condroitina/química , Dermatán Sulfato/química , Glicosaminoglicanos/metabolismo , Cofactor II de Heparina/química , Trombina/química , Animales , Biomarcadores/metabolismo , Niño , Preescolar , Ensayo de Inmunoadsorción Enzimática/métodos , Glicosaminoglicanos/orina , Humanos , Lactante , Recién Nacido , Ratones , Tamizaje Neonatal/métodos , Resultado del Tratamiento
12.
Blood Adv ; 5(1): 262-273, 2021 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-33570653

RESUMEN

Type I and III leukocyte adhesion deficiencies (LADs) are primary immunodeficiency disorders resulting in early death due to infections and additional bleeding tendency in LAD-III. The curative treatment of LAD-I and LAD-III is allogeneic hematopoietic stem cell transplantation (allo-HSCT). In this retrospective multicenter study, data were collected using the European Society for Blood and Marrow Transplantation registry; we analyzed data from 84 LAD patients from 33 centers, all receiving an allo-HSCT from 2007 to 2017. The 3-year overall survival estimate (95% confidence interval [CI]) was 83% (74-92) for the entire cohort: 84% (75-94) and 75% (50-100) for LAD-I and LAD-III, respectively. We observed cumulative incidences (95% CI) of graft failure (GF) at 3 years of 17% (9%-26%) and grade II to IV acute graft-versus-host disease (aGVHD) at 100 days of 24% (15%-34%). The estimate (95% CI) at 3 years for GF- and GVHD-II to IV-free survival as event-free survival (EFS) was 56% (46-69) for the entire cohort; 58% (46-72) and 56% (23-88) for LAD-I and LAD-III, respectively. Grade II to IV acute GVHD was a relevant risk factor for death (hazard ratio 3.6; 95% CI 1.4-9.1; P = .006). Patients' age at transplant ≥13 months, transplantation from a nonsibling donor, and any serological cytomegalovirus mismatch in donor-recipient pairs were significantly associated with severe acute GVHD and inferior EFS. The choice of busulfan- or treosulfan-based conditioning, type of GVHD prophylaxis, and serotherapy did not impact overall survival, EFS, or aGVHD. An intrinsic inflammatory component of LAD may contribute to inflammatory complications during allo-HSCT, thus providing the rationale for considering anti-inflammatory therapy pretreatment.


Asunto(s)
Enfermedad Injerto contra Huésped , Trasplante de Células Madre Hematopoyéticas , Síndrome de Deficiencia de Adhesión del Leucocito , Enfermedad Injerto contra Huésped/etiología , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Humanos , Síndrome de Deficiencia de Adhesión del Leucocito/terapia , Leucocitos , Estudios Retrospectivos
13.
Mol Genet Metab ; 99(3): 269-74, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19926322

RESUMEN

Mucopolysaccharide (MPS) diseases are lysosomal storage disorders caused by deficiencies of enzymes catabolising glycosaminoglycans (GAGs). Abnormal GAG accumulation leads to symptoms including severe progressive neurological decline, skeletal deformities, organomegally, respiratory compromise and premature death. Treatment is available for some MPS diseases; enzyme replacement therapy for MPS I, II and VI, and haematopoietic stem cell transplantation for MPS I, VI and VII. These treatments are reliant on early diagnosis of the disease and accurate monitoring of treatment outcomes. Blood enzyme levels and total urinary GAGs are commonly used biomarkers in diagnosis of MPS but are not good measures of treatment outcome. Serum heparin cofactor II-thrombin complex (HCII-T), which is a GAG regulated serpin-protease complex, has recently been identified as a promising biomarker for MPS diseases. Here we present an assessment of the HCII-T biomarker in mouse models of MPS I, IIIA and IIIB, which suggests that HCII-T is a reliable marker for MPS I when measured in serum or dried blood spots stored for over a year at 4 degrees C, but that murine MPS IIIA and IIIB cannot be reliably detected using this biomarker. We also show that HCII-T formation in vivo is dependent on the presence of excess intravenous dermatan sulphate (DS), whilst intravenous heparan sulphate (HS), does not promote complex formation effectively. This suggests that HCII-T will prove effective as a biomarker for MPS I, II, VI and VII diseases, storing dermatan sulphate but may not be as appropriate for MPS III, storing heparan sulphate. With careful sample preparation, HCII-T ELISA could prove to be a useful biomarker for both newborn screening and measurement of treatment outcomes in selected MPS diseases.


Asunto(s)
Biomarcadores/sangre , Cofactor II de Heparina/metabolismo , Mucopolisacaridosis III/diagnóstico , Mucopolisacaridosis I/diagnóstico , Trombina/metabolismo , Animales , Biomarcadores/metabolismo , Recolección de Muestras de Sangre/métodos , Dermatán Sulfato/sangre , Dermatán Sulfato/metabolismo , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Cofactor II de Heparina/análisis , Heparitina Sulfato/sangre , Heparitina Sulfato/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Mucopolisacaridosis I/sangre , Mucopolisacaridosis I/metabolismo , Mucopolisacaridosis III/sangre , Mucopolisacaridosis III/metabolismo , Trombina/análisis
14.
Mol Genet Metab ; 98(3): 235-42, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19632871

RESUMEN

Mucopolysaccharidosis type IIIB (Sanfilippo syndrome) is a lysosomal storage disease caused by a genetic defect in the production of alpha-N-acetylglucosaminidase. This results in lysosomal and extracellular accumulation of the undegraded glycosaminoglycan (GAG) substrate, heparan sulphate. Affected patients show progressive CNS degeneration characterised by mental retardation, hyperactivity and seizures, with death usually in the mid teens to early twenties. Visceral organ storage is also present but is relatively mild compared to other MPS diseases storing similar substrates. No treatments currently exist for MPS IIIB. Genistein is a broad spectrum protein tyrosine kinase inhibitor which acts on several different growth factor receptors, notably EGF and IGF receptors, both of which are important for proteoglycan synthesis. Recent work has shown that genistein can reduce GAG synthesis in patients' fibroblasts in vitro and there is evidence in patients to suggest that it may be an effective substrate reduction therapy agent for MPS III. Here we have tested the dose responses of MPS IIIB mice to daily sub-chronic dosing of genistein in half log increments compared to carrier over 8 weeks. We show clear reductions in liver lysosome compartment size in both sexes and significant dose dependent improvements in total liver GAGs and hair morphology in male MPS IIIB animals following genistein treatment. Male MPS IIIB mice exhibited considerably more liver storage than females and responded better to treatment. No changes in total GAGs, lysosomal size or reactive astrogliosis in the brain cortex were observed after 8 weeks of treatment despite evidence that genistein can cross the blood brain barrier. This is the first demonstration of genistein treatment in MPS models in vivo.


Asunto(s)
Genisteína/farmacología , Lisosomas/metabolismo , Mucopolisacaridosis III/metabolismo , Acetilglucosaminidasa/genética , Animales , Barrera Hematoencefálica/metabolismo , Modelos Animales de Enfermedad , Femenino , Hígado/metabolismo , Masculino , Ratones , Ratones Noqueados
15.
Mol Ther Methods Clin Dev ; 13: 399-413, 2019 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-31044143

RESUMEN

Hematopoietic stem cell gene therapy is a promising therapeutic strategy for the treatment of neurological disorders, since transplanted gene-corrected cells can traffic to the brain, bypassing the blood-brain barrier, to deliver therapeutic protein to the CNS. We have developed this approach for the treatment of Mucopolysaccharidosis type IIIA (MPSIIIA), a devastating lysosomal storage disease that causes progressive cognitive decline, leading to death in early adulthood. In a previous pre-clinical proof-of-concept study, we demonstrated neurological correction of MPSIIIA utilizing hematopoietic stem cell gene therapy via a lentiviral vector encoding the SGSH gene. Prior to moving to clinical trial, we have undertaken further studies to evaluate the efficiency of gene transfer into human cells and also safety studies of biodistribution and genotoxicity. Here, we have optimized hCD34+ cell transduction with clinical grade SGSH vector to provide improved pharmacodynamics and cell viability and validated effective scale-up and cryopreservation to generate an investigational medicinal product. Utilizing a humanized NSG mouse model, we demonstrate effective engraftment and biodistribution, with no vector shedding or transmission to germline cells. SGSH vector genotoxicity assessment demonstrated low transformation potential, comparable to other lentiviral vectors in the clinic. This data establishes pre-clinical safety and efficacy of HSCGT for MPSIIIA.

16.
J Clin Pathol ; 71(3): 275-278, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29055896

RESUMEN

SIFD describes a heritable, syndromic condition characterised principally by sideroblastic anaemia (SA) with immunodeficiency, fevers and developmental delay, arising in mutations within the TRNT1 gene. Other clinical manifestations of SIFD include cardiomyopathy, seizures, sensorineural hearing loss, renal dysfunction, metabolic abnormalities, hepatosplenomegaly and retinitis pigmentosa.Presentation of SIFD is variable but typically in early childhood with SA or with fever. In this report, we extend the described SIFD phenotype. We describe a kindred in which the index case presented with fetal hydrops, and early neonatal death, and the second child had severe anaemia at delivery. Both cases had prominent extramedullary erythropoiesis and numerous circulating nucleated red blood cells.


Asunto(s)
Anemia Neonatal/etiología , Anemia Sideroblástica/complicaciones , Discapacidades del Desarrollo/complicaciones , Hidropesía Fetal/etiología , Síndromes de Inmunodeficiencia/complicaciones , Hierro/metabolismo , Anemia Neonatal/patología , Anemia Sideroblástica/patología , Médula Ósea/patología , Discapacidades del Desarrollo/patología , Resultado Fatal , Femenino , Hematopoyesis Extramedular , Humanos , Hidropesía Fetal/patología , Inmunohistoquímica , Síndromes de Inmunodeficiencia/congénito , Síndromes de Inmunodeficiencia/patología , Recién Nacido , Masculino , Fenotipo
17.
Hum Gene Ther ; 27(10): 813-829, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27431943

RESUMEN

In order to support the clinical application of hematopoietic stem cell (HSC) gene therapy for mucopolysaccharidosis I (MPS I), biosafety studies were conducted to assess the toxicity and tumorigenic potential, as well as the biodistribution of HSCs and progenitor cells (HSPCs) transduced with lentiviral vectors (LV) encoding the cDNA of the alpha-iduronidase (IDUA) gene, which is mutated in MPS I patients. To this goal, toxicology and biodistribution studies were conducted, employing Good Laboratory Practice principles. Vector integration site (IS) studies were applied in order to predict adverse consequences of vector gene transfer and to obtain HSC-related information. Overall, the results obtained in these studies provided robust evidence to support the safety and tolerability of high-efficiency LV-mediated gene transfer and above-normal IDUA enzyme expression in both murine and human HSPCs and their in vivo progeny. Taken together, these investigations provide essential safety data to support clinical testing of HSC gene therapy in MPS I patients. These studies also underline criticisms associated with the use of currently available models, and highlight the value of surrogate markers of tumorigenicity that may be further explored in the future. Notably, biological evidence supporting the efficacy of gene therapy on MPS I disease and its feasibility on patients' HSCs were also generated, employing clinical-grade LVs. Finally, the clonal contribution of LV-transduced HSPCs to hematopoiesis along serial transplantation was quantified in a minimum of 200-300 clones, with the different level of repopulating cells in primary recipients being reflected in the secondary.


Asunto(s)
Terapia Genética/efectos adversos , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Iduronidasa/genética , Mucopolisacaridosis I/terapia , Animales , Regulación Enzimológica de la Expresión Génica , Técnicas de Transferencia de Gen/efectos adversos , Terapia Genética/métodos , Vectores Genéticos/efectos adversos , Células Madre Hematopoyéticas/enzimología , Humanos , Iduronidasa/efectos adversos , Iduronidasa/uso terapéutico , Lentivirus/genética , Ratones , Mucopolisacaridosis I/genética
18.
PLoS One ; 7(4): e35787, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22558223

RESUMEN

Mucopolysaccharide diseases (MPS) are caused by deficiency of glycosaminoglycan (GAG) degrading enzymes, leading to GAG accumulation. Neurodegenerative MPS diseases exhibit cognitive decline, behavioural problems and shortened lifespan. We have characterised neuropathological changes in mouse models of MPSI, IIIA and IIIB to provide a better understanding of these events.Wild-type (WT), MPSI, IIIA and IIIB mouse brains were analysed at 4 and 9 months of age. Quantitative immunohistochemistry showed significantly increased lysosomal compartment, GM2 ganglioside storage, neuroinflammation, decreased and mislocalised synaptic vesicle associated membrane protein, (VAMP2), and decreased post-synaptic protein, Homer-1, in layers II/III-VI of the primary motor, somatosensory and parietal cortex. Total heparan sulphate (HS), was significantly elevated, and abnormally N-, 6-O and 2-O sulphated compared to WT, potentially altering HS-dependent cellular functions. Neuroinflammation was confirmed by significantly increased MCP-1, MIP-1α, IL-1α, using cytometric bead arrays. An overall genotype effect was seen in all parameters tested except for synaptophysin staining, neuronal cell number and cortical thickness which were not significantly different from WT. MPSIIIA and IIIB showed significantly more pronounced pathology than MPSI in lysosomal storage, astrocytosis, microgliosis and the percentage of 2-O sulphation of HS. We also observed significant time progression of all genotypes from 4-9 months in lysosomal storage, astrocytosis, microgliosis and synaptic disorganisation but not GM2 gangliosidosis. Individual genotype*time differences were disparate, with significant progression from 4 to 9 months only seen for MPSIIIB with lysosomal storage, MPSI with astrocytocis and MPSIIIA with microgliosis as well as neuronal loss. Transmission electron microscopy of MPS brains revealed dystrophic axons, axonal storage, and extensive lipid and lysosomal storage. These data lend novel insight to MPS neuropathology, suggesting that MPSIIIA and IIIB have more pronounced neuropathology than MPSI, yet all are still progressive, at least in some aspects of neuropathology, from 4-9 months.


Asunto(s)
Lisosomas/metabolismo , Mucopolisacaridosis III/patología , Mucopolisacaridosis I/patología , Neuronas/metabolismo , Lóbulo Parietal/patología , Corteza Somatosensorial/patología , Animales , Proteínas Portadoras/biosíntesis , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Gangliósido G(M2)/biosíntesis , Glicosaminoglicanos/biosíntesis , Heparitina Sulfato/metabolismo , Proteínas de Andamiaje Homer , Inmunohistoquímica , Lisosomas/patología , Masculino , Ratones , Mucopolisacaridosis I/metabolismo , Mucopolisacaridosis III/metabolismo , Neuronas/patología , Lóbulo Parietal/metabolismo , Corteza Somatosensorial/metabolismo , Proteína 2 de Membrana Asociada a Vesículas/biosíntesis
19.
Behav Brain Res ; 209(2): 212-20, 2010 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-20138090

RESUMEN

Mucopolysaccharidosis IIIB (MPSIIIB) is a lysosomal storage disease characterised by progressive central nervous system degeneration in patients, with death usually in the late teens. Serious behavioural problems have been reported in children at the early stages of the disease, such as hyperactivity and severe sleep disturbances, which suggest alterations in circadian rhythms. We investigated the circadian rhythm of locomotor activity of young and old MPSIIIB mice, under a 24-h light-dark (LD) cycle and under constant darkness (DD), and also examined neuropeptide expression in the suprachiasmatic nucleus (SCN), site of the principal biological pacemaker. We show that MPSIIIB mice have higher activity levels during the light (resting) phase of the LD cycle, together with weaker circadian rhythms, and a longer active phase due to a late peak of activity, in both LD and DD. In addition, young MPSIIIB mice showed shorter phase delays in response to a light pulse in DD. Increased lysosomal storage, neuroinflammation and changes in the expression of Arginine Vasopressin and Vasointestinal Polypeptide, two circadian neuropeptides, were observed in the SCN, which may be in part responsible for the changes in circadian behaviour observed in MPSIIIB mice. These findings suggest an alteration of the circadian system in MPSIIIB mice, and may inform better clinical management of circadian, sleep and behavioural disturbances in patients with MPSIII.


Asunto(s)
Ritmo Circadiano/fisiología , Actividad Motora/fisiología , Mucopolisacaridosis III/fisiopatología , Núcleo Supraquiasmático/fisiopatología , Actigrafía , Factores de Edad , Análisis de Varianza , Animales , Arginina Vasopresina/metabolismo , Conducta Animal/fisiología , Modelos Animales de Enfermedad , Procesamiento de Imagen Asistido por Computador , Inmunohistoquímica , Ratones , Mucopolisacaridosis III/metabolismo , Fotoperiodo , Núcleo Supraquiasmático/metabolismo , Péptido Intestinal Vasoactivo/metabolismo
20.
PLoS One ; 5(12): e14192, 2010 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-21152017

RESUMEN

BACKGROUND: Neurodegenerative metabolic disorders such as mucopolysaccharidosis IIIB (MPSIIIB or Sanfilippo disease) accumulate undegraded substrates in the brain and are often unresponsive to enzyme replacement treatments due to the impermeability of the blood brain barrier to enzyme. MPSIIIB is characterised by behavioural difficulties, cognitive and later motor decline, with death in the second decade of life. Most of these neurodegenerative lysosomal storage diseases lack effective treatments. We recently described significant reductions of accumulated heparan sulphate substrate in liver of a mouse model of MPSIIIB using the tyrosine kinase inhibitor genistein. METHODOLOGY/PRINCIPAL FINDINGS: We report here that high doses of genistein aglycone, given continuously over a 9 month period to MPSIIIB mice, significantly reduce lysosomal storage, heparan sulphate substrate and neuroinflammation in the cerebral cortex and hippocampus, resulting in correction of the behavioural defects observed. Improvements in synaptic vesicle protein expression and secondary storage in the cerebral cortex were also observed. CONCLUSIONS/SIGNIFICANCE: Genistein may prove useful as a substrate reduction agent to delay clinical onset of MPSIIIB and, due to its multimodal action, may provide a treatment adjunct for several other neurodegenerative metabolic diseases.


Asunto(s)
Regulación de la Expresión Génica , Genisteína/farmacología , Mucopolisacaridosis III/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Neuronas/metabolismo , Animales , Barrera Hematoencefálica , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Heparitina Sulfato/química , Heterocigoto , Inmunohistoquímica/métodos , Hígado/metabolismo , Lisosomas/metabolismo , Ratones , Ratones Endogámicos C57BL , Inhibidores de Proteínas Quinasas/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA