Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
Sci Rep ; 11(1): 22593, 2021 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-34799645

RESUMEN

Mesenchymal progenitors differentiate into several tissues including bone, cartilage, and adipose. Targeting these cells in vivo is challenging, making mesenchymal progenitor cell lines valuable tools to study tissue development. Mesenchymal stem cells (MSCs) can be isolated from humans and animals; however, obtaining homogenous, responsive cells in a reproducible fashion is challenging. As such, we developed two mesenchymal progenitor cell (MPC) lines, MPC1 and MPC2, generated from bone marrow of male C57BL/6 mice. These cells were immortalized using the temperature sensitive large T-antigen, allowing for thermal control of proliferation and differentiation. Both MPC1 and MPC2 cells are capable of osteogenic, adipogenic, and chondrogenic differentiation. Under osteogenic conditions, both lines formed mineralized nodules, and stained for alizarin red and alkaline phosphatase, while expressing osteogenic genes including Sost, Fgf23, and Dmp1. Sost and Dmp1 mRNA levels were drastically reduced with addition of parathyroid hormone, thus recapitulating in vivo responses. MPC cells secreted intact (iFGF23) and C-terminal (cFGF23) forms of the endocrine hormone FGF23, which was upregulated by 1,25 dihydroxy vitamin D (1,25D). Both lines also rapidly entered the adipogenic lineage, expressing adipose markers after 4 days in adipogenic media. MPC cells were also capable of chondrogenic differentiation, displaying increased expression of cartilaginous genes including aggrecan, Sox9, and Comp. With the ability to differentiate into multiple mesenchymal lineages and mimic in vivo responses of key regulatory genes/proteins, MPC cells are a valuable model to study factors that regulate mesenchymal lineage allocation as well as the mechanisms that dictate transcription, protein modification, and secretion of these factors.


Asunto(s)
Adipocitos/citología , Técnicas de Cultivo de Célula , Condrocitos/citología , Células Madre Mesenquimatosas/citología , Osteocitos/citología , Animales , Diferenciación Celular , Línea Celular , Linaje de la Célula , Proliferación Celular , Factor-23 de Crecimiento de Fibroblastos/metabolismo , Inmunofenotipificación , Masculino , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/metabolismo
3.
Mol Genet Metab Rep ; 29: 100811, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34712574

RESUMEN

Mucopolysaccharidosis type IIIA (MPS IIIA) is characterised by a progressive neurological decline leading to early death. It is caused by bi-allelic loss-of-function mutations in SGSH encoding sulphamidase, a lysosomal enzyme required for heparan sulphate glycosaminoglycan (HS GAG) degradation, that results in the progressive build-up of HS GAGs in multiple tissues most notably the central nervous system (CNS). Skin fibroblasts from two MPS IIIA patients who presented with an intermediate and a severe clinical phenotype, respectively, were reprogrammed into induced pluripotent stem cells (iPSCs). The intermediate MPS IIIA iPSCs were then differentiated into neural progenitor cells (NPCs) and subsequently neurons. The patient derived fibroblasts, iPSCs, NPCs and neurons all displayed hallmark biochemical characteristics of MPS IIIA including reduced sulphamidase activity and increased accumulation of an MPS IIIA HS GAG biomarker. Proliferation of MPS IIIA iPSC-derived NPCs was reduced compared to control, but could be partially rescued by reintroducing functional sulphamidase enzyme, or by doubling the concentration of the mitogen fibroblast growth factor 2 (FGF2). Whilst both control heparin, and MPS IIIA HS GAGs had a similar binding affinity for FGF2, only the latter inhibited FGF signalling, suggesting accumulated MPS IIIA HS GAGs disrupt the FGF2:FGF2 receptor:HS signalling complex. Neuronal differentiation of MPS IIIA iPSC-derived NPCs was associated with a reduction in the expression of neuronal cell marker genes ßIII-TUBULIN, NF-H and NSE, revealing reduced neurogenesis compared to control. A similar result was achieved by adding MPS IIIA HS GAGs to the culture medium during neuronal differentiation of control iPSC-derived NPCs. This study demonstrates the generation of MPS IIIA iPSCs, and NPCs, the latter of which display reduced proliferation and neurogenic capacity. Reduced NPC proliferation can be explained by a model in which soluble MPS IIIA HS GAGs compete with cell surface HS for FGF2 binding. The mechanism driving reduced neurogenesis remains to be determined but appears downstream of MPS IIIA HS GAG accumulation.

4.
Curr Osteoporos Rep ; 18(6): 759-773, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33064251

RESUMEN

PURPOSE OF REVIEW: The mucopolysaccharidoses (MPS) are a group of inherited lysosomal storage disorders characterized by abnormal accumulation of glycosaminoglycans (GAGs) in cells and tissues. MPS patients frequently exhibit failures of endochondral ossification during postnatal growth leading to skeletal deformity and short stature. In this review, we outline the current understanding of the cellular and molecular mechanisms underlying failures of endochondral ossification in MPS and discuss associated treatment challenges and opportunities. RECENT FINDINGS: Studies in MPS patients and animal models have demonstrated that skeletal cells and tissues exhibit significantly elevated GAG storage from early in postnatal life and that this is associated with impaired cartilage-to-bone conversion in primary and secondary ossification centers, and growth plate dysfunction. Recent studies have begun to elucidate the underlying cellular and molecular mechanisms, including impaired chondrocyte proliferation and hypertrophy, diminished growth factor signaling, disrupted cell cycle progression, impaired autophagy, and increased cell stress and apoptosis. Current treatments such as hematopoietic stem cell transplantation and enzyme replacement therapy fail to normalize endochondral ossification in MPS. Emerging treatments including gene therapy and small molecule-based approaches hold significant promise in this regard. Failures of endochondral ossification contribute to skeletal deformity and short stature in MPS patients, increasing mortality and reducing quality of life. Early intervention is crucial for effective treatment, and there is a critical need for new approaches that normalize endochondral ossification by directly targeting affected cells and signaling pathways.


Asunto(s)
Enfermedades Óseas/etiología , Mucopolisacaridosis/complicaciones , Animales , Enfermedades Óseas/fisiopatología , Enfermedades Óseas/terapia , Trastornos del Crecimiento/etiología , Trastornos del Crecimiento/fisiopatología , Humanos , Mucopolisacaridosis/fisiopatología , Mucopolisacaridosis/terapia
5.
Mol Genet Metab Rep ; 25: 100668, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33117654

RESUMEN

Bone elongation is driven by chondrocyte proliferation and hypertrophy in the growth plate. Both processes are modulated by multiple signaling pathways including the Indian Hedgehog (IHH) signaling pathway. Mucopolysaccharidoses (MPS) are a group of lysosomal storage disorders characterized by accumulation of glycosaminoglycans (GAGs) in multiple tissues and organs, leading to a range of clinical symptoms including bone shortening through mechanisms that are not fully understood. Using MPS VII mice, we previously observed a reduction in the number of proliferating and hypertrophic chondrocytes and a reduced gene expression of Ihh in the tibial growth plate. We further demonstrate here that IHH secretion by MPS VII chondrocytes was reduced both in vitro and in vivo. While normal chondrocytes showed no response to exogenous IHH, proliferation of MPS VII chondrocytes was stimulated in response to exogenous IHH in vitro. This was accompanied by an elevated gene expression of patched receptor (Ptch1). The results from this study suggested that reduced proliferation in MPS VII growth plate may be partially due to dysfunction of the IHH signaling pathway.

6.
Mol Genet Metab ; 131(1-2): 197-205, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32739280

RESUMEN

The cause of neurodegeneration in MPS mouse models is the focus of much debate and what the underlying cause of disease pathology in MPS mice is. The timing of development of pathology and when this can be reversed or impacted is the key to developing suitable therapies in MPS. This study is the first of its kind to correlate the biochemical changes with the functional outcome as assessed using non-invasive behaviour testing across multiple mucopolysaccharidosis (MPS) mouse models. In the MPS brain, the primary lysosomal enzyme dysfunction leads to accumulation of primary glycosaminoglycans (GAGs) with gangliosides (GM2 and GM3) being the major secondary storage products. With a focus on the neuropathology, a time course experiment was conducted in MPS I, MPS IIIA, MPS VII (severe and attenuated models) in order to understand the relative timing and level of GAG and ganglioside accumulation and how this correlates to behaviour deficits. Time course analysis from 1 to 6 months of age was conducted on brain samples to assess primary GAG (uronic acid), ß-hexosaminidase enzyme activity and levels of GM2 and GM3 gangliosides. This was compared to a battery of non-invasive behaviour tests including open field, inverted grid, rotarod and water cross maze were assessed to determine effects on motor function, activity and learning ability. The results show that the GAG and ganglioside accumulation begins prior to the onset of detectable changes in learning ability and behaviour. Interestingly, the highest levels of GAG and ganglioside accumulation was observed in the MPS IIIA mouse despite having 3% residual enzyme activity. Deficits in motor function were clearly observed in the severe Gusmps/mps, which were significantly delayed in the attenuated Gustm(L175F)Sly model despite their minimal increase in detectable enzyme activity. This suggests that genotype and residual enzyme activity are not indicative of severity of disease pathology in MPS disease and there exists a window when there are considerable storage products without detectable functional deficits which may allow an alteration to occur with therapy.


Asunto(s)
Encéfalo/metabolismo , Glucuronidasa/genética , Mucopolisacaridosis III/metabolismo , Mucopolisacaridosis I/metabolismo , Mucopolisacaridosis VII/metabolismo , Animales , Encéfalo/patología , Modelos Animales de Enfermedad , Gangliósido G(M2)/genética , Gangliósido G(M2)/metabolismo , Gangliósido G(M3)/genética , Gangliósido G(M3)/metabolismo , Glicosaminoglicanos/genética , Glicosaminoglicanos/metabolismo , Heparitina Sulfato/metabolismo , Humanos , Masculino , Aprendizaje por Laberinto/fisiología , Ratones , Mucopolisacaridosis I/genética , Mucopolisacaridosis I/patología , Mucopolisacaridosis III/genética , Mucopolisacaridosis III/patología , Mucopolisacaridosis VII/genética , Mucopolisacaridosis VII/patología
7.
Bone ; 132: 115195, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31863960

RESUMEN

Endochondral bone growth is abnormal in 6 of the 11 types of mucopolysaccharidoses (MPS) disorders; resulting in short stature, reduced size of the thoracic cavity and compromised manual dexterity. Current therapies for MPS have had a limited effect on bone growth and to improve these therapies or develop adjunct approaches requires an understanding of the underlying basis of abnormal bone growth in MPS. The MPS VII mouse model replicates the reduction in long bone and vertebral length observed in human MPS. Using this model we have shown that the growth plate is elongated but contains fewer chondrocytes in the proliferative and hypertrophic zones. Endochondral bone growth is in part regulated by entry and exit from the cell cycle by growth plate chondrocytes. More MPS VII chondrocytes were positive for Ki67, a marker for active phases of the cell cycle, suggesting that more MPS VII chondrocytes were in the cell cycle. The number of cells positive for phosphorylated histone H3 was significantly reduced in MPS VII chondrocytes, suggesting fewer MPS VII chondrocytes progressed to mitotic division. While MPS VII HZ chondrocytes continued to express cyclin D1 and more cells were positive for E2F1 and phos pRb than normal, fewer MPS VII HZ chondrocytes were positive for p57kip2 a marker of terminal differentiation, suggesting fewer MPS VII chondrocytes were able to exit the cell cycle. In addition, multiple markers typical of PZ to HZ transition were not downregulated in MPS VII, in particular Sox9, Pthrpr and Wnt5a. These findings are consistent with MPS VII growth plates elongating at a slower rate than normal due to a delay in progression through the cell cycle, in particular the transition between G1 and S phases, leading to both reduced cell division and transition to the hypertrophic phenotype.


Asunto(s)
Condrocitos , Placa de Crecimiento , Animales , Ciclo Celular , Diferenciación Celular , División Celular , Hipertrofia , Ratones
8.
Mol Genet Metab ; 124(2): 135-142, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29747998

RESUMEN

Short stature is a characteristic feature of most of the mucopolysaccharidoses, a group of inherited lysosomal storage disorders caused by a single enzyme deficiency. MPS patients present with progressive skeletal defects from an early age, including short stature due to impaired cartilage-to-bone conversion (endochondral ossification). The aim of this study was to determine which murine MPS model best reproduces the bone length reduction phenotype of human MPS and use this model to determine the earliest developmental stage when disrupted endochondral ossification first appears. Gusmps/mps mice representing severe MPS VII displayed the greatest reduction in bone elongation and were chosen for histopathological analysis. Tibial development was assessed from E12.5 to 6 months of age. Chondrocytes in the region of the future primary ossification center became hypertrophic at a similar age to normal in the MPS VII mouse fetus, but a delay in bone deposition was observed with an approximate 1 day delay in the formation of the primary ossification centre. Likewise, chondrocytes in the region of the future secondary ossification center also became hypertrophic at the same age as normal in the MPS VII early postnatal mouse. Bone deposition in the secondary ossification centre was delayed by two days in the MPS VII proximal tibia (observed at postnatal day 14 (P14) compared to P12 in normal). The thickness of the tibial growth plate was larger in MPS VII mice from P9 onwards. Abnormal endochondral ossification starts in utero in MPS VII and worsens with age. It is characterized by a normal timeframe for chondrocyte hypertrophy but a delay in the subsequent deposition of bone in both the primary and secondary ossification centres, accompanied by an increase in growth plate thickness. This suggests that the signals for vascular invasion and bone deposition, some of which are derived from hypertrophic chondrocytes, are altered in MPS VII.


Asunto(s)
Enfermedades Óseas/etiología , Enfermedades Óseas/patología , Modelos Animales de Enfermedad , Mucopolisacaridosis VII/complicaciones , Osteogénesis , Tibia/patología , Animales , Animales Recién Nacidos , Diferenciación Celular , Femenino , Masculino , Ratones , Embarazo
9.
Diseases ; 5(1)2017 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-28933358

RESUMEN

Mucopolysaccharidosis type I (MPS I) is the most common form of the MPS group of genetic diseases. MPS I results from a deficiency in the lysosomal enzyme α-l-iduronidase, leading to accumulation of undegraded heparan and dermatan sulphate glycosaminoglycan (GAG) chains in patient cells. MPS children suffer from multiple organ failure and die in their teens to early twenties. In particular, MPS I children also suffer from profound mental retardation and skeletal disease that restricts growth and movement. Neither brain nor skeletal disease is adequately treated by current therapy approaches. To overcome these barriers to effective therapy we have developed and tested a treatment called substrate deprivation therapy (SDT). MPS I knockout mice were treated with weekly intravenous injections of 1 mg/kg rhodamine B for six months to assess the efficacy of SDT. Mice were assessed using biochemistry, micro-CT and a battery of behaviour tests to determine the outcome of treatment. A reduction in female bodyweight gain was observed with the treatment as well as a decrease in lung GAG. Behavioural studies showed slight improvements in inverted grid and significant improvements in learning ability for female MPS I mice treated with rhodamine B. Skeletal disease also improved with a reduction in bone mineral volume observed. Overall, rhodamine B is safe to administer to MPS I knockout mice where it had an effect on improving aspects of neurological and skeletal disease symptoms and may therefore provide a potential therapy or adjunct therapy for MPS I patients.

10.
Mol Genet Metab ; 119(3): 249-257, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27692945

RESUMEN

Severe, progressive skeletal dysplasia is a major symptom of multiple mucopolysaccharidoses (MPS) types. While a gene therapy approach initiated at birth has been shown to prevent the development of bone pathology in different animal models of MPS, the capacity to correct developed bone disease is unknown. In this study, ex vivo micro-computed tomography was used to demonstrate that bone mass and architecture of murine MPS VII L5 vertebrae were within the normal range at 1month of age but by 2months of age were significantly different to normal. The difference between normal and MPS VII BV/TV increased with age reaching a maximal difference at approximately 4months of age. In mature MPS VII bone BV/TV is increased (51.5% versus 21.5% in normal mice) due to an increase in trabecular number (6.2permm versus 3.8permm in normal mice). The total number of osteoclasts in the metaphysis of MPS VII mice was decreased, as was the percentage of osteoclasts attached to bone. MPS VII osteoblasts produced significantly more osteoprotegerin (OPG) than normal osteoblasts and supported the production of fewer osteoclasts from spleen precursor cells than normal osteoblasts in a co-culture system. In contrast, the formation of osteoclasts from MPS VII spleen monocytes was similar to normal in vitro, when exogenous RANKL and m-CSF was added to the culture medium. Administration of murine ß-glucuronidase to MPS VII mice at 4months of age, when bone disease was fully manifested, using lentiviral gene delivery resulted in a doubling of osteoclast numbers and a significant increase in attachment capacity (68% versus 29.4% in untreated MPS VII animals). Bone mineral volume rapidly decreased by 39% after gene therapy and fell within the normal range by 6months of age. Collectively, these results indicate that lentiviral-mediated gene therapy is effective in reversing established skeletal pathology in murine MPS VII.


Asunto(s)
Densidad Ósea/genética , Terapia Genética , Glucuronidasa/genética , Mucopolisacaridosis VII/terapia , Animales , Modelos Animales de Enfermedad , Técnicas de Transferencia de Gen , Glucuronidasa/administración & dosificación , Humanos , Lentivirus/genética , Ratones , Mucopolisacaridosis VII/diagnóstico por imagen , Mucopolisacaridosis VII/genética , Mucopolisacaridosis VII/patología , Osteoprotegerina/genética , Microtomografía por Rayos X
11.
Mol Genet Metab ; 118(2): 100-10, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27106513

RESUMEN

UNLABELLED: Mucopolysaccharidosis IIIA is a heritable neurodegenerative disorder resulting from the dysfunction of the lysosomal hydrolase sulphamidase. This leads to the primary accumulation of the complex carbohydrate heparan sulphate in a wide range of tissues and the secondary neuronal storage of gangliosides GM2 and GM3 in the brain. GM2 storage is associated with CNS deterioration in the GM2 gangliosidosis group of lysosomal storage disorders and may also contribute to MPS CNS disease. N-butyldeoxynojirimycin, an inhibitor of ceramide glucosyltransferase activity and therefore of ganglioside synthesis, was administered to MPS IIIA mice both prior to maximal GM2 and GM3 accumulation (early treatment) and after the maximum level of ganglioside had accumulated in the brain (late treatment) to determine if behaviour was altered by ganglioside level. Ceramide glucosyltransferase activity was decreased in both treatment groups; however, brain ganglioside levels were only decreased in the late treatment group. Learning in the water cross maze was improved in both groups and the innate fear response was also restored in both groups. A reduction in the expression of inflammatory gene Ccl3 was observed in the early treatment group, while IL1ß expression was reduced in both treatment groups. Thus, it appears that NB-DNJ elicits a transient decrease in brain ganglioside levels, some modulation of inflammatory cytokines and a functional improvement in behaviour that can be elicited both before and after overt neurological changes manifest. SYNOPSIS: NB-DNJ improves learning and restores the innate fear response in MPS IIIA mice by decreasing ceramide glucosyltransferase activity and transiently reducing ganglioside storage and/or modulating inflammatory signals.


Asunto(s)
1-Desoxinojirimicina/análogos & derivados , Inhibidores de Glicósido Hidrolasas/uso terapéutico , Mucopolisacaridosis III/tratamiento farmacológico , 1-Desoxinojirimicina/farmacología , 1-Desoxinojirimicina/uso terapéutico , Análisis de Varianza , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Quimiocina CCL3/metabolismo , Modelos Animales de Enfermedad , Miedo/efectos de los fármacos , Gangliósidos/metabolismo , Glucosiltransferasas/antagonistas & inhibidores , Glucosiltransferasas/metabolismo , Inhibidores de Glicósido Hidrolasas/farmacología , Interleucina-1beta/metabolismo , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Mucopolisacaridosis III/metabolismo , Mucopolisacaridosis III/psicología
12.
Mol Genet Metab ; 114(4): 584-93, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25748347

RESUMEN

Mucopolysaccharidoses (MPS) are inherited metabolic disorders that arise from a complete loss or a reduction in one of eleven specific lysosomal enzymes. MPS children display pathology in multiple cell types leading to tissue and organ failure and early death. Mesenchymal stem cells (MSCs) give rise to many of the cell types affected in MPS, including those that are refractory to current treatment protocols such as hematopoietic stem cell (HSC) based therapy. In this study we compared multiple MPS enzyme production by bone marrow derived (hBM) and dental pulp derived (hDP) MSCs to enzyme production by HSCs. hBM MSCs produce significantly higher levels of MPS I, II, IIIA, IVA, VI and VII enzyme than HSCs, while hDP MSCs produce significantly higher levels of MPS I, IIIA, IVA, VI and VII enzymes. Higher transfection efficiency was observed in MSCs (89%) compared to HSCs (23%) using a lentiviral vector. Over-expression of four different lysosomal enzymes resulted in up to 9303-fold and up to 5559-fold greater levels in MSC cell layer and media respectively. Stable, persistent transduction of MSCs and sustained over-expression of MPS VII enzyme was observed in vitro. Transduction of MSCs did not affect the ability of the cells to differentiate down osteogenic, adipogenic or chondrogenic lineages, but did partially delay differentiation down the non-mesodermal neurogenic lineage.


Asunto(s)
Diferenciación Celular , Glucuronidasa/biosíntesis , Células Madre Mesenquimatosas/enzimología , Mucopolisacaridosis/enzimología , Médula Ósea , Células Cultivadas , Pulpa Dental , Glucuronidasa/genética , Glicosaminoglicanos/metabolismo , Células Madre Hematopoyéticas/enzimología , Humanos , Lentivirus/genética , Mucopolisacaridosis VII/genética , Transducción Genética
13.
J Gene Med ; 16(11-12): 374-87, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25418946

RESUMEN

BACKGROUND: Mucopolysaccharidoses (MPS) are inborn metabolic disorders caused by a deficiency of glycosaminoglycan degrading enzymes. Although intravenous enzyme replacement therapy is a viable approach for the treatment of non-neuronopathic forms of MPS, its effectiveness in the central nervous system (CNS) is limited by the blood-brain barrier. Alternatively, enzyme replacement therapies and other therapies that directly target the brain represent approaches that circumvent the blood-brain barrier and, in the case of gene therapies, are intended to negate the need for repetitive dosing. METHODS: In the present study, gene therapy was targeted to the brains of young adult mice affected by mucopolysaccharidosis type IIIA (MPS IIIA) by bilateral delivery of two different therapeutic lentivirus vectors to the cerebral lateral ventricles. One vector expressed codon optimised murine sulphamidase, whereas the other co-expressed sulphamidase and sulfatase modifying factor-1. RESULTS: Six months after gene delivery, bladder distension was prevented in all treated animals, and behavioural deficits were improved. Therapeutic enzyme activity from the most efficacious vector, which was also the simpler vector, ranged from 0.5- to four-fold normal within the brains of treated animals, and the average amount of integrated vector ranged from 0.1-1 gene copies per cell. Consequently, levels of ganglioside and lysosomal ß-hexosaminidase, both of which are characteristically elevated in MPS IIIA, were significantly reduced, or were normalised. CONCLUSIONS: The present study demonstrates the efficacy of the intraventricular injection as a tool to target the brain with therapeutic genes in adult MPS IIIA mice, and provides evidence supporting this approach as a potentially effective means of treating CNS pathology in MPS IIIA patients.


Asunto(s)
Lentivirus/genética , Mucopolisacaridosis III/terapia , Animales , Encéfalo/enzimología , Encéfalo/patología , Gangliósido G(M2)/metabolismo , Gangliósido G(M3)/metabolismo , Terapia Genética , Humanos , Hidrolasas/biosíntesis , Hidrolasas/genética , Inyecciones Intraventriculares , Masculino , Aprendizaje por Laberinto , Ratones , Mucopolisacaridosis III/psicología , Transducción Genética , Resultado del Tratamiento , beta-N-Acetilhexosaminidasas/biosíntesis , beta-N-Acetilhexosaminidasas/genética
14.
Hum Gene Ther ; 25(9): 798-810, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25003807

RESUMEN

A number of mucopolysaccharidosis type VII (MPS VII) mouse models with different levels of residual enzyme activity have been created replicating the range of clinical phenotypes observed in human MPS VII patients. In this study, a lentivirus encoding murine ß-glucuronidase was administered intravenously at birth to both the severe (Gus(mps/mps) strain) and attenuated (Gus(tm(L175F)Sly) strain) mouse models of MPS VII. Circulating enzyme levels were normalized in the Gus(mps/mps) mice and were 3.5-fold higher than normal in the Gus(tm(L175F)Sly) mouse 12 and 18 months after administration. Tissue ß-glucuronidase activity increased over untreated levels in all tissues evaluated in both strains at 12 months, and the elevated level was maintained in Gus(tm(L175F)Sly) tissues at 18 months. These elevated enzyme levels reduced glycosaminoglycan storage in the liver, spleen, kidney, and heart in both models. Bone mineral volume decreased toward normal in both models after 12 months of therapy and after 18 months in the Gus(tm(L175F)Sly) mouse. Open-field exploration was improved in 18-month-old treated Gus(tm(L175F)Sly) mice, while spatial learning improved in both 12- and 18-month-old treated Gus(tm(L175F)Sly) mice. Overall, neonatal administration of lentiviral gene therapy resulted in sustained enzyme expression for up to 18 months in murine models of MPS VII. Significant improvements in biochemistry and enzymology as well as functional improvement of bone and behavior deficits in the Gus(tm(L175F)Sly) model were observed. Therapy significantly increased the lifespan of Gus(mps/mps) mice, with 12 months being the longest reported lentiviral treatment for this strain. It is important to assess the long-term outcome on enzyme levels and effect on pathology for lentiviral gene therapy to be a potential therapy for MPS patients.


Asunto(s)
Modelos Animales de Enfermedad , Terapia Genética/métodos , Glucuronidasa/sangre , Glucuronidasa/metabolismo , Mucopolisacaridosis VII/enzimología , Mucopolisacaridosis VII/terapia , Análisis de Varianza , Animales , Densidad Ósea , Conducta Exploratoria/fisiología , Vectores Genéticos/genética , Glucuronidasa/genética , Técnicas Histológicas , Riñón/metabolismo , Lentivirus , Hígado/metabolismo , Longevidad/fisiología , Ratones , Mucopolisacaridosis VII/genética , Mucopolisacaridosis VII/patología , Miocardio/metabolismo , Aprendizaje Espacial/fisiología , Especificidad de la Especie , Bazo/metabolismo
15.
Mol Genet Metab ; 106(2): 214-20, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22546219

RESUMEN

MPS disorders result from a deficiency or absence of glycosaminoglycan (GAG) degrading enzymes leading to an imbalance between the synthesis and degradation of GAGs and their subsequent accumulation in a range of cells. The inhibition of GAG synthesis using small chemical inhibitors has been proposed as a novel therapeutic approach to treatment. Several inhibitors have been shown to decrease heparan sulphate GAG synthesis and in this study we evaluated a novel fluorinated analog of N-acetylglucosamine (2-acetamido-1,3,6-tri-O-acetyl-4-deoxy-4-fluoro-D-glucopyranose (F-GlcNAc)) and rhodamine B for their ability to also inhibit the synthesis of chondroitin/dermatan and keratan sulphate GAGs present in bovine cartilage. Both inhibitors decreased GAG synthesis in chondrocyte monolayer culture and in cartilage chip explant culture in a dose dependent manner. Both inhibitors decreased the size of newly synthesised proteoglycans and in the case of F-GlcNAc this was due to a decrease in newly synthesised GAG chain size. Rhodamine B, however, did not affect GAG chain size, while both inhibitors decreased the amount of chondroitin/dermatan and keratan sulphate GAG equally. The expression of genes responsible for the initiation and elongation of chondroitin/dermatan sulphate and keratan sulphate GAGs were downregulated in the presence of rhodamine B but not in the presence of F-GlcNAc. Thus the 2 inhibitors appear to have differing effects on GAG synthesis, with F-GlcNAc inhibiting the epimerisation of UDP-GlcNAc to UDP-GalNAc thus decreasing the availability of monosaccharides for addition to the growing GAG chain, whereas rhodamine B is more likely to reduce the number of GAG chains. Together with previous data these 2 inhibitors are capable of non-specific inhibition of GAG synthesis, reducing the production of chondroitin/dermatan sulphate, keratan sulphate and heparan sulphate GAGs. As such they would be applicable to therapy in a range of MPS disorders.


Asunto(s)
Acetilglucosamina/análogos & derivados , Condrocitos/efectos de los fármacos , Condrocitos/metabolismo , Glicosaminoglicanos/biosíntesis , Rodaminas/farmacología , Acetilglucosamina/farmacología , Animales , Cartílago/metabolismo , Bovinos , Sulfatos de Condroitina/biosíntesis , Dermatán Sulfato/biosíntesis , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Sulfato de Queratano/biosíntesis , Mucopolisacaridosis/genética , Mucopolisacaridosis/metabolismo , Proteoglicanos/metabolismo
16.
Mol Genet Metab ; 106(2): 202-13, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22525091

RESUMEN

Mucopolysaccharidosis VII (MPS VII) is an autosomal recessive, lysosomal storage disorder caused by ß-glucuronidase (GUSB) deficiency, resulting in the accumulation of glycosaminoglycans (GAGs), in a variety of cell types. Severe, progressive skeletal pathology, termed dysostosis multiplex, is a prominent clinical feature of MPS VII. We have evaluated a gene therapy protocol for its efficacy in preventing the development and progression of bone pathology in MPS VII mice treated with a lentiviral vector at birth or at 7 weeks. Two weeks after injections, high levels of vector expression were observed in liver, spleen and bone marrow and to a lesser extent in kidney, lung and heart. Widespread clearance of GAG storage was observed in somatic tissues of both groups and some clearance of neuronal storage was observed in mice treated from birth. Micro-CT analysis demonstrated a significant decrease in vertebral and femoral bone mineral volume, trabecular number, bone surface density and cortical bone thickness in both treatment groups. Lumbar and femoral bone lengths were significantly decreased in untreated MPS VII mice, while growth plate heights were increased and these parameters did not change upon treatment. Small improvements in performance in the open field and rotarod behaviour tests were noted. Overall, systemic lentiviral-mediated gene therapy results in a measurable improvement in parameters of bone mass and architecture as well as biochemical and enzymatic correction. Conversely, growth plate chondrocytes were not responsive to treatment, as evidenced by the lack of improvement in vertebral and femoral bone length and growth plate height.


Asunto(s)
Terapia Genética , Vectores Genéticos , Lentivirus/genética , Mucopolisacaridosis VII/terapia , Animales , Modelos Animales de Enfermedad , Fémur/diagnóstico por imagen , Dosificación de Gen , Vectores Genéticos/administración & dosificación , Glucuronidasa/genética , Glucuronidasa/metabolismo , Placa de Crecimiento/patología , Ratones , Ratones Noqueados , Mucopolisacaridosis VII/genética , Radiografía , Columna Vertebral/diagnóstico por imagen , Distribución Tisular , Resultado del Tratamiento
17.
Curr Gene Ther ; 11(2): 132-43, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21291356

RESUMEN

Intravenous enzyme replacement therapy has been developed as a viable treatment for most of the somatic pathologies associated with the mucopolysaccharide storage disorders. However, approximately two thirds of individuals affected by a mucopolysaccharide storage disorder also display neurological disease, in these instances intravenous enzyme replacement therapy is not viable as the blood-brain barrier severely limits enzyme distribution from the peripheral circulation into the central nervous system. Accordingly, much research is now focussed on developing therapies that specifically address neurological disease, or somatic and neurological disease in combination. Therapies designed to address the underlying cause of central nervous system pathology, that is the lysosomal storage itself, can be broadly divided into two groups, those that continue the rationale of enzyme replacement, and those that address the supply side of the storage equation; that is the production of storage material. Enzyme replacement can be further divided by technology (principally direct enzyme replacement, gene replacement and cell transplantation). Here we review the current state of the art for these strategies and suggest possible future directions for research in this field. In particular, we suggest that any one approach in itself is unlikely to be as efficacious as a carefully considered combination therapy, be it a combination of some sort of enzyme replacement with substrate deprivation, or a combination of two different replacement technologies or strategies.


Asunto(s)
Terapia Genética/métodos , Mucopolisacaridosis/terapia , Enfermedades del Sistema Nervioso/terapia , Animales , Terapia de Reemplazo Enzimático , Humanos , Enfermedades por Almacenamiento Lisosomal/terapia
18.
J Gene Med ; 12(9): 717-28, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20683858

RESUMEN

BACKGROUND: The hallmark of lysosomal storage disorders (LSDs) is microscopically demonstrable lysosomal distension. In mucopolysaccharidosis type IIIA (MPS IIIA), this occurs as a result of an inherited deficiency of the lysosomal hydrolase sulphamidase. Consequently, heparan sulphate, a highly sulphated glycosaminoglycan, accumulates primarily within the cells of the reticulo-endothelial and monocyte-macrophage systems and, most importantly, neurones. Children affected by MPS IIIA experience a severe, progressive neuropathology that ultimately leads to death at around 15 years of age. METHODS: MPS IIIA pathology was addressed in a mouse model using two separate methods of therapeutic gene delivery. A lentiviral vector expressing murine sulphamidase was delivered to 6-week-old MPS IIIA affected mice either by intravenous injection, or by intraventricular infusion. Therapeutic outcomes were assessed 7 months after gene transfer. RESULTS: After intravenous gene delivery, liver sulphamidase was restored to approximately 30% of wild-type levels. The resultant widespread delivery of enzyme secreted from transduced cells to somatic tissues via the peripheral circulation corrected most somatic pathology. However, unlike an earlier study, central nervous system (CNS) pathology remained unchanged. Conversely, intraventricular gene delivery resulted in widespread sulphamidase gene delivery in (and reduced lysosomal storage throughout) the brain. Improvements in behaviour were observed in these mice, and interestingly, pathological urinary retention was prevented. CONCLUSIONS: The CNS remains the last major barrier to effective therapy for children affected by LSDs. The blood-brain barrier (BBB) limits the uptake of lysosomal enzymes from the peripheral circulation into the CNS, making direct gene delivery to the brain a reasonable, albeit more challenging, therapeutic option. Future work will further assess the relative advantages of directly targeting the brain with somatic gene delivery with sulphamidase modified to increase the efficiency of transport across the BBB.


Asunto(s)
Encéfalo/patología , Terapia Genética , Hidrolasas/genética , Lentivirus/genética , Mucopolisacaridosis III/terapia , Animales , Conducta Animal , Barrera Hematoencefálica , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Técnicas de Transferencia de Gen , Vectores Genéticos , Hidrolasas/metabolismo , Infusiones Intraventriculares , Inyecciones Intraventriculares , Hígado/enzimología , Lisosomas , Ratones , Ratones Endogámicos C57BL , Mucopolisacaridosis III/genética , Mucopolisacaridosis III/patología , beta-N-Acetilhexosaminidasas/metabolismo
19.
Mol Genet Metab ; 101(2-3): 208-13, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20650670

RESUMEN

MPS IIIA is a lysosomal storage disorder caused by mutations in the sulphamidase gene, resulting in the accumulation of heparan sulphate glycosaminoglycans (HS GAGs). Symptoms predominantly manifest in the CNS and there is no current therapy that effectively addresses neuropathology in MPS IIIA patients. Recent studies in MPS IIIA mice have shown that rhodamine B substrate deprivation therapy (SDT) (also termed substrate reduction therapy/SRT) inhibits GAG biosynthesis and, improves both somatic and CNS disease pathology. Acute overexposure to high doses of rhodamine B results in liver toxicity and is detrimental to reproductive ability. However, the long-term effects of decreasing GAG synthesis, at the low dose sufficient to alter neurological function are unknown. A trans-generational study was therefore initiated to evaluate the continuous exposure of rhodamine B treatment in MPS IIIA mice over 4 generations, including treatment during pregnancy. No alterations in litter size, liver histology or liver function were observed. Overall, there are no long-term issues with the administration of rhodamine B at the low dose tested and no adverse effects were noted during pregnancy in mice.


Asunto(s)
Glicosaminoglicanos/antagonistas & inhibidores , Hígado/efectos de los fármacos , Mucopolisacaridosis III/fisiopatología , Rodaminas/uso terapéutico , Animales , Modelos Animales de Enfermedad , Femenino , Glicosaminoglicanos/biosíntesis , Tamaño de la Camada/efectos de los fármacos , Hígado/patología , Hígado/fisiología , Ratones , Mucopolisacaridosis III/genética , Embarazo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...