Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Intervalo de año de publicación
1.
Cardiovasc Pathol ; 67: 107575, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37730078

RESUMEN

Mucopolysaccharidosis type II (MPSII) is a progressive lysosomal storage disease caused by mutations in the IDS gene, that leads to iduronate 2-sulfatase (IDS) enzyme deficiency. The enzyme catalyzes the first step of degradation of two glycosaminoglycans (GAGs), heparan sulfate (HS) and dermatan sulfate (DS). The consequences of MPSII are progressively harmful and can lead to death by cardiac failure. The aim of this study was to characterize the cardiovascular disease in MPSII mice. Thus, we evaluated the cardiovascular function of MPSII male mice at 6, 8, and 10 months of age, through functional, histological, and biochemical analyzes. Echocardiographic analyses showed a progressive loss in cardiac function, observed through parameters such as reduction in ejection fraction (46% in control versus 28% in MPS II at 10 months, P < .01) and fractional area change (31% versus 23%, P < .05). Similar results were found in parameters of vascular competence, obtained by echo Doppler. Both aortic dilatation and an increase in pulmonary resistance were observed at all time points in MPSII mice. The histological analyses showed an increase in the thickness of the heart valves (2-fold thicker than control values at 10 months). Biochemical analyzes confirmed GAG storage in these tissues, with a massive elevation of DS in the myocardium. Furthermore, an important increase in the activity of proteases such as cathepsin S and B (up to 5-fold control values) was found and could be related to the progressive loss of cardiac function observed in MPSII mice. In this work, we demonstrated that loss of cardiac function in MPSII mice started at 6 months of age, although its global cardiac capacity was still preserved at this time. Disease progressed at later time points leading to heart failure. The MPSII mice at later times reproduce many of the cardiovascular events found in patients with Hunter's disease.

2.
Adv Exp Med Biol ; 1429: 127-155, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37486520

RESUMEN

Genome editing has multiple applications in the biomedical field. They can be used to modify genomes at specific locations, being able to either delete, reduce, or even enhance gene transcription and protein expression. Here, we summarize applications of genome editing used in the field of lysosomal disorders. We focus on the development of cell lines for study of disease pathogenesis, drug discovery, and pathogenicity of specific variants. Furthermore, we highlight the main studies that use gene editing as a gene therapy platform for these disorders, both in preclinical and clinical studies. We conclude that gene editing has been able to change quickly the scenario of these disorders, allowing the development of new therapies and improving the knowledge on disease pathogenesis. Should they confirm their hype, the first gene editing-based products for lysosomal disorders could be available in the next years.


Asunto(s)
Edición Génica , Enfermedades por Almacenamiento Lisosomal , Humanos , Terapia Genética , Genoma , Enfermedades por Almacenamiento Lisosomal/genética , Enfermedades por Almacenamiento Lisosomal/terapia , Sistemas CRISPR-Cas/genética
3.
Prog Mol Biol Transl Sci ; 182: 289-325, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34175045

RESUMEN

Lysosomal disorders are a group of heterogenous diseases caused by mutations in genes that encode for lysosomal proteins. With exception of some cases, these disorders still lack both knowledge of disease pathogenesis and specific therapies. In this sense, genome editing arises as a technique that allows both the creation of specific cell lines, animal models and gene therapy protocols for these disorders. Here we explain the main applications of genome editing for lysosomal diseases, with examples based on the literature. The ability to rewrite the genome will be of extreme importance to study and potentially treat these rare disorders.


Asunto(s)
Sistemas CRISPR-Cas , Edición Génica , Animales , Terapia Genética , Genoma , Lisosomas
4.
Arch Biochem Biophys ; 709: 108970, 2021 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-34181873

RESUMEN

Glutaric acidemia type 1 (GA1) is caused by glutaryl-CoA dehydrogenase deficiency that leads to a blockage in the metabolic route of the amino acids lysine and tryptophan and subsequent accumulation of glutaric acid (GA), 3-hydroxyglutaric acids and glutarylcarnitine (C5DC). Patients predominantly manifest neurological symptoms, associated with acute striatal degeneration, as well as progressive cortical and striatum injury whose pathogenesis is not yet fully established. Current treatment includes protein/lysine restriction and l-carnitine supplementation of (L-car). The aim of this work was to evaluate behavior parameters and pro-inflammatory factors (cytokines IL-1ß, TNF-α and cathepsin-D levels), as well as the anti-inflammatory cytokine IL10 in striatum of knockout mice (Gcdh-/-) and wild type (WT) mice submitted to a normal or a high Lys diet. The potential protective effects of L-car treatment on these parameters were also evaluated. Gcdh-/- mice showed behavioral changes, including lower motor activity (decreased number of crossings) and exploratory activity (reduced number of rearings). Also, Gcdh-/- mice had significantly higher concentrations of glutarylcarnitine (C5DC) in blood and cathepsin-D (CATD), interleukin IL-1ß and tumor factor necrosis alpha (TNF-α) in striatum than WT mice. Noteworthy, L-car treatment prevented most behavioral alterations, normalized CATD levels and attenuated IL-1ß levels in striatum of Gcdh-/- mice. Finally, IL-1ß was positively correlated with CATD and C5DC levels and L-car was negatively correlated with CATD. Our results demonstrate behavioral changes and a pro-inflammatory status in striatum of the animal model of GA1 and, most importantly, L-car showed important protective effects on these alterations.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos/tratamiento farmacológico , Encefalopatías Metabólicas/tratamiento farmacológico , Carnitina/uso terapéutico , Glutaril-CoA Deshidrogenasa/deficiencia , Inflamación/tratamiento farmacológico , Fármacos Neuroprotectores/uso terapéutico , Errores Innatos del Metabolismo de los Aminoácidos/genética , Animales , Encefalopatías Metabólicas/genética , Carnitina/análogos & derivados , Carnitina/metabolismo , Catepsina D/metabolismo , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Glutaril-CoA Deshidrogenasa/genética , Aseo Animal/efectos de los fármacos , Inflamación/genética , Interleucina-1beta/metabolismo , Locomoción/efectos de los fármacos , Lisina/farmacología , Ratones Noqueados , Prueba de Campo Abierto/efectos de los fármacos , Factor de Crecimiento Transformador beta/metabolismo
5.
Metab Brain Dis ; 36(5): 1015-1027, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33620579

RESUMEN

Maple syrup urine disease (MSUD) is a genetic disorder that leads the accumulation of branched-chain amino acids (BCAA) leucine (Leu), isoleucine, valine and metabolites. The symptomatology includes psychomotor delay and mental retardation. MSUD therapy comprises a lifelong protein strict diet with low BCAA levels and is well established that high concentrations of Leu and/or its ketoacid are associated with neurological symptoms. Recently, it was demonstrated that the phenylbutyrate (PBA) have the ability to decrease BCAA concentrations. This work aimed the development of lipid-based nanoparticles loaded with PBA, capable of targeting to the central nervous system in order to verify its action mechanisms on oxidative stress and cell death in brain of rats subjected to a MSUD chronic model. PBA-loaded nanoparticles treatment was effective in significantly decreasing BCAA concentration in plasma and Leu in the cerebral cortex of MSUD animals. Furthermore, PBA modulate the activity of catalase, superoxide dismutase, glutathione peroxidase and glutathione reductase enzymes, as well as preventing the oxidative damage to lipid membranes and proteins. PBA was also able to decrease the glial fibrillary acidic protein concentrations and partially decreased the reactive species production and caspase-3 activity in MSUD rats. Taken together, the data indicate that the PBA-loaded nanoparticles could be an efficient adjuvant in the MSUD therapy, protecting against oxidative brain damage and neuroinflammation.


Asunto(s)
Aminoácidos de Cadena Ramificada/sangre , Corteza Cerebral/efectos de los fármacos , Enfermedad de la Orina de Jarabe de Arce/metabolismo , Nanopartículas/administración & dosificación , Estrés Oxidativo/efectos de los fármacos , Fenilbutiratos/administración & dosificación , Animales , Catalasa/metabolismo , Corteza Cerebral/metabolismo , Glutatión Peroxidasa/metabolismo , Enfermedad de la Orina de Jarabe de Arce/sangre , Enfermedad de la Orina de Jarabe de Arce/inducido químicamente , Ratas , Ratas Wistar , Superóxido Dismutasa/metabolismo
6.
Interdiscip Sci ; 13(1): 34-43, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33475959

RESUMEN

Mucopolysaccharidoses are caused by a deficiency of enzymes involved in the degradation of glycosaminoglycans. Heart diseases are a significant cause of morbidity and mortality in MPS patients, even in conditions in which enzyme replacement therapy is available. In this sense, cardiovascular manifestations, such as heart hypertrophy, cardiac function reduction, increased left ventricular chamber, and aortic dilatation, are among the most frequent. However, the downstream events which influence the heart dilatation process are unclear. Here, we employed systems biology tools together with transcriptomic data to investigate new elements that may be involved in aortic dilatation in Mucopolysaccharidoses syndrome. We identified candidate genes involved in biological processes related to inflammatory responses, deposition of collagen, and lipid accumulation in the cardiovascular system that may be involved in aortic dilatation in the Mucopolysaccharidoses I and VII. Furthermore, we investigated the molecular mechanisms of losartan treatment in Mucopolysaccharidoses I mice to underscore how this drug acts to prevent aortic dilation. Our data indicate that the association between the TGF-b signaling pathway, Fos, and Col1a1 proteins can play an essential role in aortic dilation's pathophysiology and its subsequent improvement by losartan treatment.


Asunto(s)
Mucopolisacaridosis , Animales , Dilatación , Terapia de Reemplazo Enzimático , Glicosaminoglicanos/uso terapéutico , Humanos , Ratones , Mucopolisacaridosis/tratamiento farmacológico , Transcriptoma
7.
J Inherit Metab Dis ; 44(3): 740-750, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33145772

RESUMEN

Mucopolysaccharidosis type I (MPS I) is a lysosomal storage disorder caused by mutations in the IDUA gene, that codifies the alpha-L-iduronidase enzyme, which deficiency leads to storage of glycosaminoglycans, with multiple clinical manifestations. One of the leading causes of death in MPS I patients are cardiac complications such as cardiac valve thickening, conduction abnormalities, myocardial dysfunction, and cardiac hypertrophy. The mechanism leading to cardiac dysfunction in MPS I is not entirely understood. In a previous study, we have demonstrated that losartan and propranolol improved the cardiac function in MPS I mice. Thus, we aimed to investigate whether the pathways influenced by these drugs may modulate the cardiac remodeling process in MPS I mice. According to our previous observation, losartan and propranolol restore the heart function, without altering valve thickness. MPS I mice presented reduced activation of AKT and ERK1/2, increased activity of cathepsins, but no alteration in metalloproteinase activity was observed. Animals treated with losartan showed a reduction in cathepsin activity and restored ERK1/2 activation. While both losartan and propranolol improved heart function, no mechanistic evidence was found for propranolol so far. Our results suggest that losartan or propranolol could be used to ameliorate the cardiac disease in MPS I and could be considered as adjuvant treatment candidates for therapy optimization.


Asunto(s)
Cardiopatías/patología , Losartán/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Mucopolisacaridosis I/tratamiento farmacológico , Remodelación Ventricular/efectos de los fármacos , Animales , Modelos Animales de Enfermedad , Ecocardiografía , Femenino , Cardiopatías/tratamiento farmacológico , Cardiopatías/genética , Iduronidasa/genética , Sistema de Señalización de MAP Quinasas/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Mucopolisacaridosis I/genética , Mucopolisacaridosis I/patología , Mutación
8.
BMC Cancer ; 20(1): 474, 2020 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-32456685

RESUMEN

BACKGROUND: Chemotherapeutics can stimulate immune antitumor response by inducing immunogenic cell death (ICD), which is activated by Damage-Associated Molecular Patterns (DAMPs) like the exposure of calreticulin (CRT) on the cell surface, the release of ATP and the secretion of High Mobility Group Box 1 (HMGB1). METHODS: Here, we investigated the levels of ICD-associated DAMPs induced by chemotherapeutics commonly used in the clinical practice of non-small cell lung cancer (NSCLC) and the association of these DAMPs with apoptosis and autophagy. A549 human lung adenocarcinoma cells were treated with clinically relevant doses of cisplatin, carboplatin, etoposide, paclitaxel and gemcitabine. We assessed ICD-associated DAMPs, cell viability, apoptosis and autophagy in an integrated way. RESULTS: Cisplatin and its combination with etoposide induced the highest levels of apoptosis, while etoposide was the less pro-apoptotic treatment. Cisplatin also induced the highest levels of ICD-associated DAMPs, which was not incremented by co-treatments. Etoposide induced the lower levels of ICD and the highest levels of autophagy, suggesting that the cytoprotective role of autophagy is dominant in relation to its pro-ICD role. High levels of CRT were associated with better prognosis in TCGA databank. In an integrative analysis we found a strong positive correlation between DAMPs and apoptosis, and a negative correlation between cell number and ICD-associated DAMPs as well as between autophagy and apoptosis markers. We also purpose a mathematical integration of ICD-associated DAMPs in an index (IndImunnog) that may represent with greater biological relevance this process. Cisplatin-treated cells showed the highest IndImmunog, while etoposide was the less immunogenic and the more pro-autophagic treatment. CONCLUSIONS: Cisplatin alone induced the highest levels of ICD-associated DAMPs, so that its combination with immunotherapy may be a promising therapeutic strategy in NSCLC.


Asunto(s)
Adenocarcinoma del Pulmón/metabolismo , Alarminas/metabolismo , Antineoplásicos/farmacología , Muerte Celular Inmunogénica , Neoplasias Pulmonares/metabolismo , Células A549 , Adenocarcinoma del Pulmón/tratamiento farmacológico , Adenocarcinoma del Pulmón/mortalidad , Adenocarcinoma del Pulmón/patología , Adenosina Trifosfato/metabolismo , Alarminas/efectos de los fármacos , Apoptosis , Autofagia , Calreticulina/metabolismo , Carboplatino/farmacología , Caspasa 3/metabolismo , Supervivencia Celular , Cisplatino/farmacología , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Etopósido/farmacología , Proteína HMGB1/metabolismo , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Paclitaxel/farmacología , Pronóstico , Gemcitabina
9.
Int J Mol Sci ; 21(4)2020 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-32093427

RESUMEN

Mucopolysaccharidosis type I (MPS I) is caused by genetic deficiency of α-l-iduronidase and impairment of lysosomal catabolism of heparan sulfate and dermatan sulfate. In the brain, these substrates accumulate in the lysosomes of neurons and glial cells, leading to neuroinflammation and neurodegeneration. Their storage also affects lysosomal homeostasis-inducing activity of several lysosomal proteases including cathepsin B (CATB). In the central nervous system, increased CATB activity has been associated with the deposition of amyloid plaques due to an alternative pro-amyloidogenic processing of the amyloid precursor protein (APP), suggesting a potential role of this enzyme in the neuropathology of MPS I. In this study, we report elevated levels of protein expression and activity of CATB in cortex tissues of 6-month-old MPS I (Idua -/- mice. Besides, increased CATB leakage from lysosomes to the cytoplasm of Idua -/- cortical pyramidal neurons was indicative of damaged lysosomal membranes. The increased CATB activity coincided with an elevated level of the 16-kDa C-terminal APP fragment, which together with unchanged levels of ß-secretase 1 was suggestive for the role of this enzyme in the amyloidogenic APP processing. Neuronal accumulation of Thioflavin-S-positive misfolded protein aggregates and drastically increased levels of neuroinflammatory glial fibrillary acidic protein (GFAP)-positive astrocytes and CD11b-positive activated microglia were observed in Idua -/- cortex by confocal fluorescent microscopy. Together, our results point to the existence of a novel CATB-associated alternative amyloidogenic pathway in MPS I brain induced by lysosomal storage and potentially leading to neurodegeneration.


Asunto(s)
Precursor de Proteína beta-Amiloide/metabolismo , Catepsina B/metabolismo , Corteza Cerebral/metabolismo , Mucopolisacaridosis I/metabolismo , Células Piramidales/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animales , Astrocitos/metabolismo , Astrocitos/patología , Catepsina B/genética , Corteza Cerebral/patología , Proteína Ácida Fibrilar de la Glía/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Lisosomas/metabolismo , Lisosomas/patología , Ratones , Ratones Noqueados , Mucopolisacaridosis I/genética , Mucopolisacaridosis I/patología , Células Piramidales/patología
10.
Am J Med Genet A ; 182(3): 469-483, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31926052

RESUMEN

Several studies have been published on the frequency of the mucopolysaccharidoses (MPS) in different countries. The objective of the present study was to estimate the birth prevalence (BP) of MPS in Brazil. MPS diagnosis registered at MPS-Brazil Network and in Instituto Vidas Raras were reviewed. BP was estimated by (a) the number of registered patients born between 1994 and 2015 was divided by the number of live births (LBs), and (b) a sample of 1,000 healthy individuals was tested for the most frequent variant in IDUA gene in MPS I (p.Trp402Ter) to estimate the frequency of heterozygosity and homozygosity. (a) The BP based on total number of LBs was (cases per 100,000 LBs): MPS overall: 1.25; MPS I: 0.24; MPS II: 0.37; MPS III: 0.21; MPS IV: 0.14; MPS VI: 0.28; MPS VII: 0.02. (b) The overall frequency of p.Trp402Ter was 0.002. Considering the frequency of heterozygotes for the p.Trp402Ter IDUA variant in the RS state, the frequency of this variant among MPS I patients and the relative frequency of the different MPSs, we estimated the birth prevalence of MPS in total and of each MPS type, as follows: MPS overall: 4.62; MPS I: 0.95; MPS II: 1.32; MPS III: 0.56; MPS IV: 0.57; MPS VI: 1.02; MPS VII: 0.05. This study provided original data about BP and relative frequency of the MPS types, in Brazil, based on the frequency of the commonest IDUA pathogenic variant and in the records of two large patient databases.


Asunto(s)
Iduronidasa/genética , Mucopolisacaridosis/genética , Brasil/epidemiología , Femenino , Humanos , Iduronidasa/sangre , Nacimiento Vivo , Masculino , Mucopolisacaridosis/sangre , Mucopolisacaridosis/epidemiología , Mucopolisacaridosis/patología , Mucopolisacaridosis I/sangre , Mucopolisacaridosis I/epidemiología , Mucopolisacaridosis I/genética , Mucopolisacaridosis II/sangre , Mucopolisacaridosis II/epidemiología , Mucopolisacaridosis II/genética , Mucopolisacaridosis III/sangre , Mucopolisacaridosis III/epidemiología , Mucopolisacaridosis III/genética , Mucopolisacaridosis VI/sangre , Mucopolisacaridosis VI/epidemiología , Mucopolisacaridosis VI/genética , Mutación/genética
11.
Gene Ther ; 27(1-2): 74-84, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31827259

RESUMEN

Mucopolysaccharidosis type I (MPS I) is caused by deficiency of alpha-L-iduronidase (IDUA), leading to multisystemic accumulation of glycosaminoglycans (GAG). Untreated MPS I patients may die in the first decades of life, mostly due to cardiovascular and respiratory complications. We previously reported that the treatment of newborn MPS I mice with intravenous administration of lipossomal CRISPR/Cas9 complexes carrying the murine Idua gene aiming at the ROSA26 locus resulted in long-lasting IDUA activity and GAG reduction in various tissues. Following this, the present study reports the effects of gene editing in cardiovascular, respiratory, bone, and neurologic functions in MPS I mice. Bone morphology, specifically the width of zygomatic and femoral bones, showed partial improvement. Although heart valves were still thickened, cardiac mass and aortic elastin breaks were reduced, with normalization of aortic diameter. Pulmonary resistance was normalized, suggesting improvement in respiratory function. In contrast, behavioral abnormalities and neuroinflammation still persisted, suggesting deterioration of the neurological functions. The set of results shows that gene editing performed in newborn animals improved some manifestations of the MPS I disorder in bone, respiratory, and cardiovascular systems. However, further studies will be imperative to find better delivery strategies to reach "hard-to-treat" tissues to ensure better systemic and neurological effects.


Asunto(s)
Iduronidasa/genética , Mucopolisacaridosis I/terapia , ARN no Traducido/genética , Animales , Animales Recién Nacidos , Enfermedades Óseas/genética , Sistemas CRISPR-Cas/genética , Sistema Cardiovascular/metabolismo , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Modelos Animales de Enfermedad , Femenino , Edición Génica/métodos , Terapia Genética/métodos , Glicosaminoglicanos/metabolismo , Iduronidasa/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Mucopolisacaridosis I/genética , Mucopolisacaridosis I/fisiopatología , Fenotipo , ARN no Traducido/metabolismo
12.
J Control Release ; 288: 23-33, 2018 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-30170069

RESUMEN

Mucopolysaccharidosis type I (MPS I) is a multisystemic disorder caused by the deficiency of alpha-L-iduronidase (IDUA) that leads to intracellular accumulation of glycosaminoglycans (GAG). In the present study we aimed to use cationic liposomes carrying the CRISPR/Cas9 plasmid and a donor vector for in vitro and in vivo MPS I gene editing, and compare to treatment with naked plasmids. The liposomal formulation was prepared by microfluidization. Complexes were obtained by the addition of DNA at +4/-1 charge ratio. The overall results showed complexes of about 110 nm, with positive zeta potential of +30 mV. The incubation of the complexes with fibroblasts from MPS I patients led to a significant increase in IDUA activity and reduction of lysosomal abnormalities. Hydrodynamic injection of the liposomal complex in newborn MPS I mice led to a significant increase in serum IDUA levels for up to six months. The biodistribution of complexes after hydrodynamic injection was markedly detected in the lungs and heart, corroborating the results of increased IDUA activity and decreased GAG storage especially in these tissues, while the group that received the naked plasmids presented increased enzyme activity especially in the liver. Furthermore, animals treated with the liposomal formulation presented improvement in cardiovascular parameters, one of the main causes of death observed in MPS I patients. We conclude that the IDUA production in multiple organs had a significant beneficial effect on the characteristics of MPS I disease, which may bring hope to gene therapy of Hurler patients.


Asunto(s)
Edición Génica , Terapia Genética , Mucopolisacaridosis I/genética , Mucopolisacaridosis I/terapia , Animales , Sistemas CRISPR-Cas , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Femenino , Fibroblastos/efectos de los fármacos , Humanos , Iduronidasa/metabolismo , Liposomas , Masculino , Ratones Endogámicos C57BL , Mucopolisacaridosis I/metabolismo , Distribución Tisular
13.
Life Sci ; 196: 102-109, 2018 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-29366749

RESUMEN

Mucopolysaccharidosis type I (MPS I) is a lysosomal storage disorder with multisystemic features, including heart enlargement, heart valve dysfunction, and aortic stiffness and dilatation. Previous studies have shown that MPS I mice overexpress cathepsin B (CtsB) in multiple tissues, including those from the cardiovascular system. Here, we hypothesized that inhibition of CtsB could ameliorate cardiac function parameters, as well as aorta and valve abnormalities found in MPS I. First, we found that total elastase activity in an MPS I aorta is elevated. Following that, we demonstrated that CtsB leaks from the lysosome in MPS I human fibroblasts, possibly acting as a degradative agent of extracellular matrix components from the aorta, cardiac muscle, and heart valves. We then used a CtsB inhibitor in vivo in the MPS I mouse model. After 4 months of treatment, partial inhibition of CtsB activity in treated mice reduced aortic dilatation, as well as heart valve thickening, and led to improvements in cardiac function parameters, although none of these were completely normalized. Based on these results, we conclude that lysosomal alterations in this disease promote leakage of CtsB to outside the organelle, where this protein can have multiple pathological roles. CtsB inhibition improved cardiovascular parameters in MPS I mice and can have a potential benefit in this disease.


Asunto(s)
Sistema Cardiovascular/patología , Catepsina B/antagonistas & inhibidores , Inhibidores de Cisteína Proteinasa/uso terapéutico , Dipéptidos/uso terapéutico , Mucopolisacaridosis I/diagnóstico por imagen , Mucopolisacaridosis I/tratamiento farmacológico , Animales , Aorta/patología , Aorta/fisiopatología , Sistema Cardiovascular/diagnóstico por imagen , Catepsina B/metabolismo , Colagenasas/metabolismo , Femenino , Fibroblastos/metabolismo , Pruebas de Función Cardíaca , Enfermedades de las Válvulas Cardíacas/diagnóstico por imagen , Enfermedades de las Válvulas Cardíacas/tratamiento farmacológico , Enfermedades de las Válvulas Cardíacas/patología , Humanos , Lisosomas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Mucopolisacaridosis I/patología , Elastasa Pancreática/metabolismo
15.
J. inborn errors metab. screen ; 5: e160028, 2017. tab
Artículo en Inglés | LILACS-Express | LILACS | ID: biblio-1090937

RESUMEN

Abstract Lysosomal storage disorders (LSDs) are a group of diseases with multisystemic features. Current treatments have limitations and gene therapy arises as a promising treatment option. Here, we discuss some of the most recent studies for gene therapy in LSD, vectors used, and outcomes. In particular, the approaches used in animal models aiming to correct the central nervous system, the eye, and the bones are highlighted. Finally, we discuss the recent reports of clinical trials using this technology for these diseases. We conclude that gene therapy for LSD has gathered a substantial amount of evidence from animal models to know its potential and limitations. First evidences from clinical trials using both adeno-associated and lentiviral vectors show that this approach is safe and efficient and therefore could provide an effective treatment for several LSD in the near future.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...