Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
JCI Insight ; 6(16)2021 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-34197342

RESUMEN

Functional loss of myosin Vb (MYO5B) induces a variety of deficits in intestinal epithelial cell function and causes a congenital diarrheal disorder, microvillus inclusion disease (MVID). The impact of MYO5B loss on differentiated cell lineage choice has not been investigated. We quantified the populations of differentiated epithelial cells in tamoxifen-induced, epithelial cell-specific MYO5B-knockout (VilCreERT2 Myo5bfl/fl) mice utilizing digital image analysis. Consistent with our RNA-sequencing data, MYO5B loss induced a reduction in tuft cells in vivo and in organoid cultures. Paneth cells were significantly increased by MYO5B deficiency along with expansion of the progenitor cell zone. We further investigated the effect of lysophosphatidic acid (LPA) signaling on epithelial cell differentiation. Intraperitoneal LPA significantly increased tuft cell populations in both control and MYO5B-knockout mice. Transcripts for Wnt ligands were significantly downregulated by MYO5B loss in intestinal epithelial cells, whereas Notch signaling molecules were unchanged. Additionally, treatment with the Notch inhibitor dibenzazepine (DBZ) restored the populations of secretory cells, suggesting that the Notch pathway is maintained in MYO5B-deficient intestine. MYO5B loss likely impairs progenitor cell differentiation in the small intestine in vivo and in vitro, partially mediated by Wnt/Notch imbalance. Notch inhibition and/or LPA treatment may represent an effective therapeutic approach for treatment of MVID.


Asunto(s)
Síndromes de Malabsorción/genética , Microvellosidades/patología , Mucolipidosis/genética , Miosina Tipo V/deficiencia , Receptores Notch/metabolismo , Vía de Señalización Wnt/genética , Animales , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Células Cultivadas , Dibenzazepinas/farmacología , Modelos Animales de Enfermedad , Enterocitos/efectos de los fármacos , Enterocitos/metabolismo , Humanos , Mucosa Intestinal/citología , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/patología , Yeyuno/citología , Yeyuno/efectos de los fármacos , Yeyuno/patología , Lisofosfolípidos/farmacología , Lisofosfolípidos/uso terapéutico , Síndromes de Malabsorción/tratamiento farmacológico , Síndromes de Malabsorción/patología , Ratones , Ratones Noqueados , Microvellosidades/genética , Mucolipidosis/tratamiento farmacológico , Mucolipidosis/patología , Miosina Tipo V/genética , Organoides , Cultivo Primario de Células , Receptores Notch/antagonistas & inhibidores , Células Madre/fisiología , Vía de Señalización Wnt/efectos de los fármacos
2.
J Biol Chem ; 295(39): 13556-13569, 2020 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-32727849

RESUMEN

Mutations in the galactosidase ß 1 (GLB1) gene cause lysosomal ß-galactosidase (ß-Gal) deficiency and clinical onset of the neurodegenerative lysosomal storage disease, GM1 gangliosidosis. ß-Gal and neuraminidase 1 (NEU1) form a multienzyme complex in lysosomes along with the molecular chaperone, protective protein cathepsin A (PPCA). NEU1 is deficient in the neurodegenerative lysosomal storage disease sialidosis, and its targeting to and stability in lysosomes strictly depend on PPCA. In contrast, ß-Gal only partially depends on PPCA, prompting us to investigate the role that ß-Gal plays in the multienzyme complex. Here, we demonstrate that ß-Gal negatively regulates NEU1 levels in lysosomes by competitively displacing this labile sialidase from PPCA. Chronic cellular uptake of purified recombinant human ß-Gal (rhß-Gal) or chronic lentiviral-mediated GLB1 overexpression in GM1 gangliosidosis patient fibroblasts coincides with profound secondary NEU1 deficiency. A regimen of intermittent enzyme replacement therapy dosing with rhß-Gal, followed by enzyme withdrawal, is sufficient to augment ß-Gal activity levels in GM1 gangliosidosis patient fibroblasts without promoting NEU1 deficiency. In the absence of ß-Gal, NEU1 levels are elevated in the GM1 gangliosidosis mouse brain, which are restored to normal levels following weekly intracerebroventricular dosing with rhß-Gal. Collectively, our results highlight the need to carefully titrate the dose and dosing frequency of ß-Gal augmentation therapy for GM1 gangliosidosis. They further suggest that intermittent intracerebroventricular enzyme replacement therapy dosing with rhß-Gal is a tunable approach that can safely augment ß-Gal levels while maintaining NEU1 at physiological levels in the GM1 gangliosidosis brain.


Asunto(s)
Terapia de Reemplazo Enzimático , Fibroblastos/enzimología , Lisosomas/enzimología , Mucolipidosis , beta-Galactosidasa/uso terapéutico , Animales , Células CHO , Cricetulus , Humanos , Lisosomas/genética , Ratones , Ratones Mutantes , Mucolipidosis/tratamiento farmacológico , Mucolipidosis/enzimología , Mucolipidosis/genética , Neuraminidasa/genética , Neuraminidasa/metabolismo
3.
Gastroenterology ; 159(4): 1390-1405.e20, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32534933

RESUMEN

BACKGROUND & AIM: Myosin VB (MYO5B) is an essential trafficking protein for membrane recycling in gastrointestinal epithelial cells. The inactivating mutations of MYO5B cause the congenital diarrheal disease, microvillus inclusion disease (MVID). MYO5B deficiency in mice causes mislocalization of SGLT1 and NHE3, but retained apical function of CFTR, resulting in malabsorption and secretory diarrhea. Activation of lysophosphatidic acid (LPA) receptors can improve diarrhea, but the effect of LPA on MVID symptoms is unclear. We investigated whether LPA administration can reduce the epithelial deficits in MYO5B-knockout mice. METHODS: Studies were conducted with tamoxifen-induced, intestine-specific knockout of MYO5B (VilCreERT2;Myo5bflox/flox) and littermate controls. Mice were given LPA, an LPAR2 agonist (GRI977143), or vehicle for 4 days after a single injection of tamoxifen. Apical SGLT1 and CFTR activities were measured in Üssing chambers. Intestinal tissues were collected, and localization of membrane transporters was evaluated by immunofluorescence analysis in tissue sections and enteroids. RNA sequencing and enrichment analysis were performed with isolated jejunal epithelial cells. RESULTS: Daily administration of LPA reduced villus blunting, frequency of multivesicular bodies, and levels of cathepsins in intestinal tissues of MYO5B-knockout mice compared with vehicle administration. LPA partially restored the brush border height and the localization of SGLT1 and NHE3 in small intestine of MYO5B-knockout mice and enteroids. The SGLT1-dependent short-circuit current was increased and abnormal CFTR activities were decreased in jejunum from MYO5B-knockout mice given LPA compared with vehicle. CONCLUSIONS: LPA may regulate a MYO5B-independent trafficking mechanism and brush border maturation, and therefore be developed for treatment of MVID.


Asunto(s)
Lisofosfolípidos/uso terapéutico , Síndromes de Malabsorción/tratamiento farmacológico , Síndromes de Malabsorción/patología , Microvellosidades/patología , Mucolipidosis/tratamiento farmacológico , Mucolipidosis/patología , Miosina Tipo V/deficiencia , Transportador 1 de Sodio-Glucosa/metabolismo , Animales , Modelos Animales de Enfermedad , Enterocitos/patología , Síndromes de Malabsorción/etiología , Ratones , Ratones Noqueados , Mucolipidosis/etiología
4.
Curr Pharm Des ; 24(24): 2870-2875, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30198428

RESUMEN

Sialic acid residues that make part of the cell surface repertoire of carbohydrate residues are implicated in various physiological processes and human pathologies. Sialidases, or neuraminidases, are the enzymes that are able to cleave and release the sialic acid residues, while trans-sialidases can transfer the residues from donor to acceptor molecules. They are important for processing the surface glycolipids and glycoproteins. Therapeutic potential of pharmacological sialidases inhibition is currently actively studied. Knowledge and expertise gained from genetic defects leading to human sialidase deficiency can be used for designing such drugs. In this review, we discuss the current progress in studying sialidases and their inhibitors and the relevance of these studies to developing novel therapeutic approaches. In vitro studies suggest that some sialidase inhibitors might be useful therapeutics for treating sialidosis, cancer, infections, immune diseases, atherosclerosis and other pathologies. Consequently, there is a field for further research and development. A thorough investigation of human sialidases is therefore crucial to human health.


Asunto(s)
Aterosclerosis/tratamiento farmacológico , Inhibidores Enzimáticos/farmacología , Glicoproteínas/antagonistas & inhibidores , Enfermedades del Sistema Inmune/tratamiento farmacológico , Mucolipidosis/tratamiento farmacológico , Neoplasias/tratamiento farmacológico , Neuraminidasa/antagonistas & inhibidores , Aterosclerosis/metabolismo , Inhibidores Enzimáticos/química , Glicoproteínas/metabolismo , Humanos , Enfermedades del Sistema Inmune/metabolismo , Mucolipidosis/metabolismo , Neoplasias/metabolismo , Neuraminidasa/metabolismo , Ácidos Siálicos/química , Ácidos Siálicos/metabolismo
5.
Hum Mol Genet ; 27(15): 2725-2738, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29771310

RESUMEN

Mucolipidosis IV (MLIV) is an orphan neurodevelopmental disease that causes severe neurologic dysfunction and loss of vision. Currently there is no therapy for MLIV. It is caused by loss of function of the lysosomal channel mucolipin-1, also known as TRPML1. Knockout of the Mcoln1 gene in a mouse model mirrors clinical and neuropathologic signs in humans. Using this model, we previously observed robust activation of microglia and astrocytes in early symptomatic stages of disease. Here we investigate the consequence of mucolipin-1 loss on astrocyte inflammatory activation in vivo and in vitro and apply a pharmacologic approach to restore Mcoln1-/- astrocyte homeostasis using a clinically approved immunomodulator, fingolimod. We found that Mcoln1-/- mice over-express numerous pro-inflammatory cytokines, some of which were also over-expressed in astrocyte cultures. Changes in the cytokine profile in Mcoln1-/- astrocytes are concomitant with changes in phospho-protein signaling, including activation of PI3K/Akt and MAPK pathways. Fingolimod promotes cytokine homeostasis, down-regulates signaling within the PI3K/Akt and MAPK pathways and restores the lysosomal compartment in Mcoln1-/- astrocytes. These data suggest that fingolimod is a promising candidate for preclinical evaluation in our MLIV mouse model, which, in case of success, can be rapidly translated into clinical trial.


Asunto(s)
Astrocitos/efectos de los fármacos , Astrocitos/patología , Encéfalo/efectos de los fármacos , Clorhidrato de Fingolimod/farmacología , Mucolipidosis/patología , Animales , Encéfalo/metabolismo , Encéfalo/patología , Células Cultivadas , Citocinas/metabolismo , Modelos Animales de Enfermedad , Encefalitis/tratamiento farmacológico , Encefalitis/genética , Encefalitis/metabolismo , Encefalitis/patología , Femenino , Regulación de la Expresión Génica , Proteínas de Membrana de los Lisosomas/metabolismo , Masculino , Ratones Noqueados , Mucolipidosis/tratamiento farmacológico , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfoproteínas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Canales de Potencial de Receptor Transitorio/genética , Canales de Potencial de Receptor Transitorio/metabolismo
8.
J Neurochem ; 133(2): 174-86, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25581256

RESUMEN

A growing body of evidence indicates that valproic acid (VPA), a histone deacetylase inhibitor used to treat epilepsy and mood disorders, has histone deacetylase-related and -unrelated neurotoxic activity, the mechanism of which is still poorly understood. We report that VPA induces neuronal cell death through an atypical calpain-dependent necroptosis pathway that initiates with downstream activation of c-Jun N-terminal kinase 1 (JNK1) and increased expression of receptor-interacting protein 1 (RIP-1) and is accompanied by cleavage and mitochondrial release/nuclear translocation of apoptosis-inducing factor, mitochondrial release of Smac/DIABLO, and inhibition of the anti-apoptotic protein X-linked inhibitor of apoptosis (XIAP). Coinciding with apoptosis-inducing factor nuclear translocation, VPA induces phosphorylation of the necroptosis-associated histone H2A family member H2AX, which is known to contribute to lethal DNA degradation. These signals are inhibited in neuronal cells that express constitutively activated MEK/ERK and/or PI3-K/Akt survival pathways, allowing them to resist VPA-induced cell death. The data indicate that VPA has neurotoxic activity and identify a novel calpain-dependent necroptosis pathway that includes JNK1 activation and RIP-1 expression. A growing body of evidence indicates that valproic acid (VPA) has neurotoxic activity, the mechanism of which is still poorly understood. We report, for the first time, that VPA activates a previously unrecognized calpain-dependent necroptosis cascade that initiates with JNK1 activation and involves AIF cleavage/nuclear translocation and H2AX phosphorylation as well as an altered Smac/DIABLO to XIAP balance.


Asunto(s)
Calpaína/farmacología , Inhibidores Enzimáticos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Neuronas/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Ácido Valproico/farmacología , Animales , Apoptosis/efectos de los fármacos , Calpaína/metabolismo , Muerte Celular/efectos de los fármacos , Línea Celular , Relación Dosis-Respuesta a Droga , Activación Enzimática/efectos de los fármacos , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Mucolipidosis/tratamiento farmacológico , Necrosis/inducido químicamente , Factor de Crecimiento Nervioso/farmacología , Células PC12 , Proteínas Serina-Treonina Quinasas/metabolismo , Ratas , Proteína Serina-Treonina Quinasas de Interacción con Receptores , Factores de Tiempo
9.
Am J Physiol Gastrointest Liver Physiol ; 307(10): G992-G1001, 2014 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-25258405

RESUMEN

Microvillus inclusion disease (MVID) is an autosomal recessive condition resulting in intractable secretory diarrhea in newborns due to loss-of-function mutations in myosin Vb (Myo5b). Previous work suggested that the apical recycling endosomal (ARE) compartment is the primary location for phosphoinositide-dependent protein kinase 1 (PDK1) signaling. Because the ARE is disrupted in MVID, we tested the hypothesis that polarized signaling is affected by Myo5b dysfunction. Subcellular distribution of PDK1 was analyzed in human enterocytes from MVID/control patients by immunocytochemistry. Using Myo5b knockdown (kd) in Caco-2BBe cells, we studied phosphorylated kinases downstream of PDK1, electrophysiological parameters, and net water flux. PDK1 was aberrantly localized in human MVID enterocytes and Myo5b-deficient Caco-2BBe cells. Two PDK1 target kinases were differentially affected: phosphorylated atypical protein kinase C (aPKC) increased fivefold and phosohoprotein kinase B slightly decreased compared with control. PDK1 redistributed to a soluble (cytosolic) fraction and copurified with basolateral endosomes in Myo5b kd. Myo5b kd cells showed a decrease in net water absorption that could be reverted with PDK1 inhibitors. We conclude that, in addition to altered apical expression of ion transporters, depolarization of PDK1 in MVID enterocytes may lead to aberrant activation of downstream kinases such as aPKC. The findings in this work suggest that PDK1-dependent signaling may provide a therapeutic target for treating MVID.


Asunto(s)
Polaridad Celular , Enterocitos/metabolismo , Síndromes de Malabsorción/metabolismo , Microvellosidades/patología , Mucolipidosis/metabolismo , Cadenas Pesadas de Miosina/metabolismo , Miosina Tipo V/metabolismo , Transducción de Señal , Proteínas Quinasas Dependientes de 3-Fosfoinosítido/antagonistas & inhibidores , Proteínas Quinasas Dependientes de 3-Fosfoinosítido/metabolismo , Células CACO-2 , Estudios de Casos y Controles , Regulación hacia Abajo , Endosomas/metabolismo , Enterocitos/efectos de los fármacos , Humanos , Síndromes de Malabsorción/tratamiento farmacológico , Síndromes de Malabsorción/genética , Microvellosidades/genética , Microvellosidades/metabolismo , Terapia Molecular Dirigida , Mucolipidosis/tratamiento farmacológico , Mucolipidosis/genética , Mutación , Cadenas Pesadas de Miosina/genética , Miosina Tipo V/genética , Fosforilación , Proteína Quinasa C/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Transporte de Proteínas , Proteínas Proto-Oncogénicas c-akt/metabolismo , Interferencia de ARN , Transducción de Señal/efectos de los fármacos , Transfección , Agua/metabolismo
12.
J Clin Neurosci ; 20(2): 324-5, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23157844

RESUMEN

Levetiracetam can suppress sialidosis-related myoclonus but its effects on cerebral metabolism warrant elucidation. In this report, fluorodeoxyglucose-positron emission tomography was conducted before and after levetiracetam in a sialidosis patient. By subtracting the drug "off" from "on" signals, regions of enhanced metabolism were shown to be allocated mostly in the bilateral fronto-temporal regions whereas regions of reduced metabolism were distributed mainly in the occipital areas. Imaging changes suggest that the effects of levetiracetam may be different in different brain regions. The drug may also elicit fronto-temporal inhibitory impulses to suppress the vigorous myoclonus in sialidosis.


Asunto(s)
Corteza Cerebral/diagnóstico por imagen , Mucolipidosis/diagnóstico por imagen , Mucolipidosis/tratamiento farmacológico , Mioclonía/diagnóstico por imagen , Mioclonía/tratamiento farmacológico , Piracetam/análogos & derivados , Adulto , Corteza Cerebral/efectos de los fármacos , Humanos , Levetiracetam , Masculino , Neuroimagen/métodos , Piracetam/farmacología , Piracetam/uso terapéutico , Tomografía de Emisión de Positrones/métodos
16.
Z Rheumatol ; 69(6): 527-38, 2010 Aug.
Artículo en Alemán | MEDLINE | ID: mdl-20532791

RESUMEN

Lysosomal storage diseases are a heterogeneous group of disorders caused by lysosomal enzyme dysfunction. Individually they are very rare, but this group as a whole has a prevalence of more than 1:8,000 live births. While severe phenotypes are easily diagnosed this can be a real challenge with attenuated forms. Because musculoskeletal complaints are frequently the first reason for the patient to seek medical advice, the rheumatologist plays a key role for the early recognition of these diseases. Since several of these can be treated very effectively by enzyme replacement, a timely diagnosis and start of therapy are essential to avoid irreversible organ damage and poor quality of life. Therefore, each clinical rheumatologist should be aware of the cardinal symptoms of lysosomal storage diseases.


Asunto(s)
Enfermedades por Almacenamiento Lisosomal/diagnóstico , Enfermedades Raras/diagnóstico , Adolescente , Adulto , Niño , Conducta Cooperativa , Diagnóstico Diferencial , Terapia de Reemplazo Enzimático , Enfermedad de Fabry/diagnóstico , Enfermedad de Fabry/tratamiento farmacológico , Enfermedad de Fabry/genética , Enfermedad de Gaucher/diagnóstico , Enfermedad de Gaucher/tratamiento farmacológico , Enfermedad de Gaucher/genética , Enfermedad del Almacenamiento de Glucógeno/diagnóstico , Enfermedad del Almacenamiento de Glucógeno/tratamiento farmacológico , Enfermedad del Almacenamiento de Glucógeno/genética , Humanos , Comunicación Interdisciplinaria , Enfermedades por Almacenamiento Lisosomal/tratamiento farmacológico , Enfermedades por Almacenamiento Lisosomal/genética , Mucolipidosis/diagnóstico , Mucolipidosis/tratamiento farmacológico , Mucolipidosis/genética , Mucopolisacaridosis/diagnóstico , Mucopolisacaridosis/tratamiento farmacológico , Mucopolisacaridosis/genética , Fenotipo , Enfermedades Raras/tratamiento farmacológico , Enfermedades Raras/genética , Esfingolipidosis/diagnóstico , Esfingolipidosis/tratamiento farmacológico , Esfingolipidosis/genética , Adulto Joven
17.
Mol Genet Metab ; 85(3): 181-9, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15979029

RESUMEN

Given the success of enzyme replacement therapy (ERT) in treating the systemic manifestations in a number of lysosomal storage disorders (LSDs), we evaluated the effect of ERT on the mouse model of sialidosis. This glycoproteinosis, which affects primarily the reticuloendothelial (RE) system, is caused by deficiency of lysosomal neuraminidase (NEU1) and consequent accumulation of sialylated glycoconjugates. NEU1 lacks a functional mannose-6-phosphate recognition marker and is not endocytosed by mammalian cells. However, the enzyme produced in insect cells has features that allow its effective uptake by RE cells and macrophages via the mannose receptor, and therefore represent an alternative method of therapy. In this study we tested the therapeutic efficacy of baculovirus (BV) expressed mouse neuraminidase (Neu1) in sialidosis mice. Four-week-old Neu1-/- mice were first injected intravenously with a single dose of the recombinant enzyme for assessment of the half-life of mannosylated Neu1 in vivo. Afterwards, a short-term ERT with a total of five enzyme injections over a 2-week period was performed for evaluation of phenotype correction. Neu1 infused alone or co-administered with its associated protein, protective protein/cathepsin A (PPCA) was effectively taken up by resident macrophages in many tissues. Restored Neu1 activity persisted for up to 4 days, depending on the tissue, and resulted in a significant reduction of lysosomal storage. However, beyond 2 weeks of treatment, ERT mice developed a severe immune response towards the exogenous Neu1 enzyme. These results may have important implications for ERT in sialidosis patients.


Asunto(s)
Catepsina A/uso terapéutico , Mucolipidosis/tratamiento farmacológico , Neuraminidasa/uso terapéutico , Animales , Baculoviridae/enzimología , Modelos Animales de Enfermedad , Quimioterapia Combinada , Marcación de Gen , Humanos , Ratones , Ratones Mutantes , Mucolipidosis/enzimología , Neuraminidasa/genética , Neuraminidasa/inmunología , Neuraminidasa/metabolismo , Proteínas Recombinantes/uso terapéutico
19.
Biochem Biophys Res Commun ; 168(2): 479-84, 1990 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-2334418

RESUMEN

There is evidence that lysosomal proteases mediate the intracellular degradation of structurally abnormal collagen. I-Cell disease (Mucolipidosis II) is characterized by marked deficiency of many lysosomal hydrolases, including the collagenolytic enzyme cathepsin B. The experiments reported here tested the hypothesis that degradation of abnormal collagen would be severely impaired in I-cells. Skin fibroblasts from 3 patients with I-cell disease were incubated with and without cis-hydroxyproline, a proline analog that causes structural abnormalities in collagen, and [14C]proline. The amount of [14C]hydroxyproline in a low molecular weight fraction relative to total [14C]hydroxyproline was used as a measure of intracellular collagen degradation. Levels of degradation were significantly higher in I-cells exposed to cis-hydroxyproline than in cells incubated without the analog. Similar data were obtained for normal human fetal lung fibroblasts incubated under the same conditions. Degradation of [125I]-epidermal growth factor was used to assess the functionality of the lysosomal pathway for protein degradation, and it was much lower in I-cells than in normal cells. It can be concluded that a completely functional complement of lysosomal enzymes is not necessary for structurally abnormal collagen to be degraded intracellularly; the data suggest that a nonlysosomal pathway exists.


Asunto(s)
Colágeno/metabolismo , Hidroxiprolina/farmacología , Mucolipidosis/metabolismo , Células Cultivadas , Factor de Crecimiento Epidérmico/farmacología , Fibroblastos , Humanos , Masculino , Mucolipidosis/tratamiento farmacológico , Mucolipidosis/patología , Desnaturalización Proteica
20.
Pediatr Radiol ; 20(1-2): 80-4, 1989.
Artículo en Inglés | MEDLINE | ID: mdl-2602022

RESUMEN

Evolution of the early bone lesions in two children with mucolipidosis 2 was followed from birth. The progression of the bone changes did not differ from healing of rickets. Low levels of 1,25-(OH)2-D3 were found in one child and he was treated with vitamin D; resolution of the rachitic changes was more rapid than in the untreated child. It is suggested that in mucolipidosis 2 bone reacts to two independent factors, one controlling calcium metabolism, the other depending on the primary lysosomal enzyme defect. Since ricket-like features are not present in the other mucolipidoses or mucopolysaccharidoses, the defect of calcium metabolism seems to be related to the specific enzyme defect of mucolipidosis 2.


Asunto(s)
Huesos/diagnóstico por imagen , Mucolipidosis/tratamiento farmacológico , Vitamina D/uso terapéutico , Envejecimiento/metabolismo , Enfermedades Óseas Metabólicas/congénito , Enfermedades Óseas Metabólicas/diagnóstico por imagen , Huesos/metabolismo , Humanos , Recién Nacido , Masculino , Mucolipidosis/complicaciones , Mucolipidosis/diagnóstico por imagen , Osteoporosis/diagnóstico por imagen , Osteoporosis/etiología , Radiografía
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA