Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Am Soc Nephrol ; 32(6): 1389-1408, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33785583

RESUMEN

BACKGROUND: Podocytes embrace the glomerular capillaries with foot processes, which are interconnected by a specialized adherens junction to ultimately form the filtration barrier. Altered adhesion and loss are common features of podocyte injury, which could be mediated by shedding of cell-adhesion molecules through the regulated activity of cell surface-expressed proteases. A Disintegrin and Metalloproteinase 10 (ADAM10) is such a protease known to mediate ectodomain shedding of adhesion molecules, among others. Here we evaluate the involvement of ADAM10 in the process of antibody-induced podocyte injury. METHODS: Membrane proteomics, immunoblotting, high-resolution microscopy, and immunogold electron microscopy were used to analyze human and murine podocyte ADAM10 expression in health and kidney injury. The functionality of ADAM10 ectodomain shedding for podocyte development and injury was analyzed, in vitro and in vivo, in the anti-podocyte nephritis (APN) model in podocyte-specific, ADAM10-deficient mice. RESULTS: ADAM10 is selectively localized at foot processes of murine podocytes and its expression is dispensable for podocyte development. Podocyte ADAM10 expression is induced in the setting of antibody-mediated injury in humans and mice. Podocyte ADAM10 deficiency attenuates the clinical course of APN and preserves the morphologic integrity of podocytes, despite subepithelial immune-deposit formation. Functionally, ADAM10-related ectodomain shedding results in cleavage of the cell-adhesion proteins N- and P-cadherin, thus decreasing their injury-related surface levels. This favors podocyte loss and the activation of downstream signaling events through the Wnt signaling pathway in an ADAM10-dependent manner. CONCLUSIONS: ADAM10-mediated ectodomain shedding of injury-related cadherins drives podocyte injury.


Asunto(s)
Proteína ADAM10/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Proteínas de la Membrana/metabolismo , Nefritis/metabolismo , Síndrome Nefrótico/metabolismo , Podocitos/metabolismo , Podocitos/patología , Insuficiencia Renal Crónica/metabolismo , Proteína ADAM10/genética , Secretasas de la Proteína Precursora del Amiloide/genética , Animales , Autoanticuerpos/efectos adversos , Nitrógeno de la Urea Sanguínea , Cadherinas/metabolismo , Adhesión Celular , Comunicación Celular , Membrana Celular/metabolismo , Células Cultivadas , Creatinina/orina , Modelos Animales de Enfermedad , Femenino , Barrera de Filtración Glomerular/patología , Barrera de Filtración Glomerular/fisiopatología , Humanos , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Nefritis/patología , Síndrome Nefrótico/patología , Podocitos/fisiología , Proteómica , Análisis de Matrices Tisulares , Transcriptoma , Vía de Señalización Wnt
2.
EMBO Rep ; 20(3)2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30733280

RESUMEN

Signal peptide peptidase (SPP) and the four homologous SPP-like (SPPL) proteases constitute a family of intramembrane aspartyl proteases with selectivity for type II-oriented transmembrane segments. Here, we analyse the physiological function of the orphan protease SPPL2c, previously considered to represent a non-expressed pseudogene. We demonstrate proteolytic activity of SPPL2c towards selected tail-anchored proteins. Despite shared ER localisation, SPPL2c and SPP exhibit distinct, though partially overlapping substrate spectra and inhibitory profiles, and are organised in different high molecular weight complexes. Interestingly, SPPL2c is specifically expressed in murine and human testis where it is primarily localised in spermatids. In mice, SPPL2c deficiency leads to a partial loss of elongated spermatids and reduced motility of mature spermatozoa, but preserved fertility. However, matings of male and female SPPL2c-/- mice exhibit reduced litter sizes. Using proteomics we identify the sarco/endoplasmic reticulum Ca2+-ATPase (SERCA2)-regulating protein phospholamban (PLN) as a physiological SPPL2c substrate. Accumulation of PLN correlates with a decrease in intracellular Ca2+ levels in elongated spermatids that likely contribute to the compromised male germ cell differentiation and function of SPPL2c-/- mice.


Asunto(s)
Ácido Aspártico Endopeptidasas/metabolismo , Proteínas de Unión al Calcio/metabolismo , Membrana Celular/enzimología , Células Germinativas/metabolismo , Proteínas de la Membrana/metabolismo , Secuencia de Aminoácidos , Animales , Ácido Aspártico Endopeptidasas/química , Calcio/metabolismo , Retículo Endoplásmico/metabolismo , Femenino , Células HEK293 , Células HeLa , Homeostasis , Humanos , Masculino , Proteínas de la Membrana/química , Ratones , Especificidad de Órganos , Espermátides/metabolismo , Especificidad por Sustrato , Testículo/enzimología
3.
Biochem J ; 477(17): 3433-3451, 2020 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-32856704

RESUMEN

Mucopolysaccharidoses comprise a group of rare metabolic diseases, in which the lysosomal degradation of glycosaminoglycans (GAGs) is impaired due to genetically inherited defects of lysosomal enzymes involved in GAG catabolism. The resulting intralysosomal accumulation of GAG-derived metabolites consequently manifests in neurological symptoms and also peripheral abnormalities in various tissues like liver, kidney, spleen and bone. As each GAG consists of differently sulfated disaccharide units, it needs a specific, but also partly overlapping set of lysosomal enzymes to accomplish their complete degradation. Recently, we identified and characterized the lysosomal enzyme arylsulfatase K (Arsk) exhibiting glucuronate-2-sulfatase activity as needed for the degradation of heparan sulfate (HS), chondroitin sulfate (CS) and dermatan sulfate (DS). In the present study, we investigated the physiological relevance of Arsk by means of a constitutive Arsk knockout mouse model. A complete lack of glucuronate desulfation was demonstrated by a specific enzyme activity assay. Arsk-deficient mice show, in an organ-specific manner, a moderate accumulation of HS and CS metabolites characterized by 2-O-sulfated glucuronate moieties at their non-reducing ends. Pathophysiological studies reflect a rather mild phenotype including behavioral changes. Interestingly, no prominent lysosomal storage pathology like bone abnormalities were detected. Our results from the Arsk mouse model suggest a new although mild form of mucopolysacharidose (MPS), which we designate MPS type IIB.


Asunto(s)
Arilsulfatasas/metabolismo , Sulfatos de Condroitina/metabolismo , Heparitina Sulfato/metabolismo , Mucopolisacaridosis/metabolismo , Animales , Arilsulfatasas/genética , Sulfatos de Condroitina/genética , Activación Enzimática , Heparitina Sulfato/genética , Ratones , Ratones Noqueados , Mucopolisacaridosis/genética
4.
J Immunol ; 199(1): 172-185, 2017 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-28550201

RESUMEN

The invariant chain (CD74) mediates assembly and targeting of MHC class II (MHCII) complexes. In endosomes, CD74 undergoes sequential degradation by different proteases, including cathepsin S (CatS) and the intramembrane protease signal peptide peptidase-like 2a (SPPL2a). In their absence, CD74 N-terminal fragments (NTFs) accumulate. In SPPL2a-/- B cells, such an NTF impairs endosomal trafficking and BCR signal transduction. In mice, this leads to a loss of splenic B cells beyond the transitional stage 1. To gain insight into CD74 determinants and the role of MHCII, we compared B cells from CatS-/- , SPPL2a-/- , and SPPL2a-MHCII double-deficient mice. We assessed differentiation of B cells in bone marrow and spleen and analyzed their endosomal morphology, BCR expression, and signal transduction. We demonstrate that MHCII is dispensable for the B cell phenotype of SPPL2a-/- mice, further supporting a CD74-intrinsic effect. Despite significant vacuolization of endosomal compartments similar to SPPL2a-/- B cells, CatS-/- traditional stage 1 B cells show unimpaired degradation of endocytic cargo, have intact BCR signaling, and do not exhibit any relevant defects in maturation. This could indicate that CD74 NTF-induced structural changes of endosomes are not directly involved in these processes. We further found that the block of CD74 degradation in CatS-/- B cells is incomplete, so that NTF levels are significantly lower than in SPPL2a-/- B cells. This suggests a dose dependency and threshold for the CD74 NTF-associated impairment of B cell signaling and maturation. In addition, different functional properties of the longer, MHCII-bound CD74 NTF could contribute to the milder phenotype of CatS-/- B cells.


Asunto(s)
Antígenos de Diferenciación de Linfocitos B/inmunología , Linfocitos B/inmunología , Genes MHC Clase II , Antígenos de Histocompatibilidad Clase II/inmunología , Animales , Antígenos de Diferenciación de Linfocitos B/metabolismo , Ácido Aspártico Endopeptidasas/deficiencia , Ácido Aspártico Endopeptidasas/genética , Ácido Aspártico Endopeptidasas/metabolismo , Catepsinas/deficiencia , Catepsinas/genética , Catepsinas/metabolismo , Diferenciación Celular , Endosomas/inmunología , Endosomas/fisiología , Antígenos de Histocompatibilidad Clase II/metabolismo , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Transducción de Señal
5.
J Biol Chem ; 291(8): 4128-43, 2016 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-26719331

RESUMEN

The intercalated disc (ID) is a "hot spot" for heart disease, as several ID proteins have been found mutated in cardiomyopathy. Myozap is a recent addition to the list of ID proteins and has been implicated in serum-response factor signaling. To elucidate the cardiac consequences of targeted deletion of myozap in vivo, we generated myozap-null mutant (Mzp(-/-)) mice. Although Mzp(-/-) mice did not exhibit a baseline phenotype, increased biomechanical stress due to pressure overload led to accelerated cardiac hypertrophy, accompanied by "super"-induction of fetal genes, including natriuretic peptides A and B (Nppa/Nppb). Moreover, Mzp(-/-) mice manifested a severe reduction of contractile function, signs of heart failure, and increased mortality. Expression of other ID proteins like N-cadherin, desmoplakin, connexin-43, and ZO-1 was significantly perturbed upon pressure overload, underscored by disorganization of the IDs in Mzp(-/-) mice. Exploration of the molecular causes of enhanced cardiac hypertrophy revealed significant activation of ß-catenin/GSK-3ß signaling, whereas MAPK and MKL1/serum-response factor pathways were inhibited. In summary, myozap is required for proper adaptation to increased biomechanical stress. In broader terms, our data imply an essential function of the ID in cardiac remodeling beyond a mere structural role and emphasize the need for a better understanding of this molecular structure in the context of heart disease.


Asunto(s)
Cardiomegalia/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Sistema de Señalización de MAP Quinasas , Proteínas Musculares/metabolismo , Factor de Respuesta Sérica/metabolismo , Transactivadores/metabolismo , beta Catenina/metabolismo , Animales , Cardiomegalia/genética , Cardiomegalia/patología , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3 beta , Ratones , Ratones Noqueados , Proteínas Musculares/genética , Ratas , Factor de Respuesta Sérica/genética , Transactivadores/genética , Factores de Transcripción , beta Catenina/genética
6.
Hum Mol Genet ; 24(7): 1856-68, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25452429

RESUMEN

Deficiency of arylsulfatase G (ARSG) leads to a lysosomal storage disease in mice resembling biochemical and pathological features of the mucopolysaccharidoses and particularly features of mucopolysaccharidosis type III (Sanfilippo syndrome). Here we show that Arsg KO mice share common neuropathological findings with other Sanfilippo syndrome models and patients, but they can be clearly distinguished by the limitation of most phenotypic alterations to the cerebellum, presenting with ataxia as the major neurological finding. We determined in detail the expression of ARSG in the central nervous system and observed highest expression in perivascular macrophages (which are characterized by abundant vacuolization in Arsg KO mice) and oligodendrocytes. To gain insight into possible mechanisms leading to ataxia, the pathology in older adult mice (>12 months) was investigated in detail. This study revealed massive loss of Purkinje cells and gliosis in the cerebellum, and secondary accumulation of glycolipids like GM2 and GM3 gangliosides and unesterified cholesterol in surviving Purkinje cells, as well as neurons of some other brain regions. The abundant presence of ubiquitin and p62-positive aggregates in degenerating Purkinje cells coupled with the absence of significant defects in macroautophagy is consistent with lysosomal membrane permeabilization playing a role in the pathogenesis of Arsg-deficient mice and presumably Sanfilippo disease in general. Our data delineating the phenotype of mucopolysaccharidosis IIIE in a mouse KO model should help in the identification of possible human cases of this disease.


Asunto(s)
Arilsulfatasas/deficiencia , Ataxia/enzimología , Mucopolisacaridosis III/enzimología , Animales , Arilsulfatasas/genética , Ataxia/genética , Ataxia/metabolismo , Ataxia/patología , Cerebelo/citología , Cerebelo/metabolismo , Modelos Animales de Enfermedad , Femenino , Gliosis/metabolismo , Glucolípidos/metabolismo , Humanos , Masculino , Ratones , Ratones Noqueados , Mucopolisacaridosis III/genética , Mucopolisacaridosis III/metabolismo , Mucopolisacaridosis III/patología , Células de Purkinje/metabolismo
7.
Proc Natl Acad Sci U S A ; 111(43): 15573-8, 2014 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-25316793

RESUMEN

Mutations within the lysosomal enzyme ß-glucocerebrosidase (GC) result in Gaucher disease and represent a major risk factor for developing Parkinson disease (PD). Loss of GC activity leads to accumulation of its substrate glucosylceramide and α-synuclein. Since lysosomal activity of GC is tightly linked to expression of its trafficking receptor, the lysosomal integral membrane protein type-2 (LIMP-2), we studied α-synuclein metabolism in LIMP-2-deficient mice. These mice showed an α-synuclein dosage-dependent phenotype, including severe neurological impairments and premature death. In LIMP-2-deficient brains a significant reduction in GC activity led to lipid storage, disturbed autophagic/lysosomal function, and α-synuclein accumulation mediating neurotoxicity of dopaminergic (DA) neurons, apoptotic cell death, and inflammation. Heterologous expression of LIMP-2 accelerated clearance of overexpressed α-synuclein, possibly through increasing lysosomal GC activity. In surviving DA neurons of human PD midbrain, LIMP-2 levels were increased, probably to compensate for lysosomal GC deficiency. Therefore, we suggest that manipulating LIMP-2 expression to increase lysosomal GC activity is a promising strategy for the treatment of synucleinopathies.


Asunto(s)
Glucosilceramidasa/metabolismo , Proteínas de Membrana de los Lisosomas/metabolismo , alfa-Sinucleína/metabolismo , Animales , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Tronco Encefálico/efectos de los fármacos , Tronco Encefálico/enzimología , Tronco Encefálico/patología , Tronco Encefálico/ultraestructura , Gliosis/complicaciones , Gliosis/patología , Humanos , Lípidos/química , Proteínas de Membrana de los Lisosomas/deficiencia , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Lisosomas/patología , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Neuronas/ultraestructura , Neurotoxinas/toxicidad
8.
Proc Natl Acad Sci U S A ; 109(26): 10310-5, 2012 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-22689975

RESUMEN

Deficiency of glycosaminoglycan (GAG) degradation causes a subclass of lysosomal storage disorders called mucopolysaccharidoses (MPSs), many of which present with severe neuropathology. Critical steps in the degradation of the GAG heparan sulfate remain enigmatic. Here we show that the lysosomal arylsulfatase G (ARSG) is the long-sought glucosamine-3-O-sulfatase required to complete the degradation of heparan sulfate. Arsg-deficient mice accumulate heparan sulfate in visceral organs and the central nervous system and develop neuronal cell death and behavioral deficits. This accumulated heparan sulfate exhibits unique nonreducing end structures with terminal N-sulfoglucosamine-3-O-sulfate residues, allowing diagnosis of the disorder. Recombinant human ARSG is able to cleave 3-O-sulfate groups from these residues as well as from an authentic 3-O-sulfated N-sulfoglucosamine standard. Our results demonstrate the key role of ARSG in heparan sulfate degradation and strongly suggest that ARSG deficiency represents a unique, as yet unknown form of MPS, which we term MPS IIIE.


Asunto(s)
Arilsulfatasas/antagonistas & inhibidores , Mucopolisacaridosis/etiología , Sulfatasas/metabolismo , Animales , Conducta Animal , Ratones , Mucopolisacaridosis/enzimología
9.
Development ; 138(3): 495-505, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21205794

RESUMEN

The disintegrin and metalloproteinase Adam10 has been implicated in the regulation of key signaling pathways that determine skin morphogenesis and homeostasis. To address the in vivo relevance of Adam10 in the epidermis, we have selectively disrupted Adam10 during skin morphogenesis and in adult skin. K14-Cre driven epidermal Adam10 deletion leads to perinatal lethality, barrier impairment and absence of sebaceous glands. A reduction of spinous layers, not associated with differences in either proliferation or apoptosis, indicates that loss of Adam10 triggers a premature differentiation of spinous keratinocytes. The few surviving K14-Adam10-deleted mice and mice in which Adam10 was deleted postnatally showed loss of hair, malformed vibrissae, epidermal hyperproliferation, cyst formation, thymic atrophy and upregulation of the cytokine thymic stromal lymphopoetin (TSLP), thus indicating non cell-autonomous multi-organ disease resulting from a compromised barrier. Together, these phenotypes closely resemble skin specific Notch pathway loss-of-function phenotypes. Notch processing is indeed strongly reduced resulting in decreased levels of Notch intracellular domain fragment and functional Notch signaling. The data identify Adam10 as the major Site-2 processing enzyme for Notch in the epidermis in vivo, and thus as a central regulator of skin development and maintenance.


Asunto(s)
Proteínas ADAM/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Células Epidérmicas , Epidermis/metabolismo , Proteínas de la Membrana/metabolismo , Receptores Notch/metabolismo , Proteínas ADAM/genética , Proteína ADAM10 , Secretasas de la Proteína Precursora del Amiloide/genética , Animales , Western Blotting , Proliferación Celular , Células Cultivadas , Inmunohistoquímica , Queratinocitos/citología , Queratinocitos/metabolismo , Proteínas de la Membrana/genética , Ratones , Ratones Mutantes , Análisis de Secuencia por Matrices de Oligonucleótidos , Receptores Notch/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/genética , Transducción de Señal/fisiología
10.
Front Mol Biosci ; 10: 1169658, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37342207

RESUMEN

Despite the identification of numerous molecular pathways modulating cardiac hypertrophy its pathogenesis is not completely understood. In this study we define an unexpected role for Fibin ("fin bud initiation factor homolog") in cardiomyocyte hypertrophy. Via gene expression profiling in hypertrophic murine hearts after transverse aortic constriction we found a significant induction of Fibin. Moreover, Fibin was upregulated in another mouse model of cardiac hypertrophy (calcineurin-transgenics) as well as in patients with dilated cardiomyopathy. Immunoflourescence microscopy revealed subcellular localization of Fibin at the sarcomeric z-disc. Overexpression of Fibin in neonatal rat ventricular cardiomyocytes revealed a strong anti-hypertrophic effect through inhibiting both, NFAT- and SRF-dependent signalling. In contrast, transgenic mice with cardiac-restricted overexpression of Fibin developed dilated cardiomyopathy, accompanied by induction of hypertrophy-associated genes. Moreover, Fibin overexpression accelerated the progression to heart failure in the presence of prohypertrophic stimuli such as pressure overload and calcineurin overexpression. Histological and ultrastructural analyses surprisingly showed large protein aggregates containing Fibin. On the molecular level, aggregate formation was accompanied by an induction of the unfolded protein response subsequent UPR-mediated apoptosis and autophagy. Taken together, we identified Fibin as a novel potent negative regulator of cardiomyocyte hypertrophy in vitro. Yet, heart-specific Fibin overexpression in vivo causes development of a protein-aggregate-associated cardiomyopathy. Because of close similarities to myofibrillar myopathies, Fibin represents a candidate gene for cardiomyopathy and Fibin transgenic mice may provide additional mechanistic insight into aggregate formation in these diseases.

11.
Am J Hum Genet ; 82(3): 673-84, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18308289

RESUMEN

Action myoclonus-renal failure syndrome (AMRF) is an autosomal-recessive disorder with the remarkable combination of focal glomerulosclerosis, frequently with glomerular collapse, and progressive myoclonus epilepsy associated with storage material in the brain. Here, we employed a novel combination of molecular strategies to find the responsible gene and show its effects in an animal model. Utilizing only three unrelated affected individuals and their relatives, we used homozygosity mapping with single-nucleotide polymorphism chips to localize AMRF. We then used microarray-expression analysis to prioritize candidates prior to sequencing. The disorder was mapped to 4q13-21, and microarray-expression analysis identified SCARB2/Limp2, which encodes a lysosomal-membrane protein, as the likely candidate. Mutations in SCARB2/Limp2 were found in all three families used for mapping and subsequently confirmed in two other unrelated AMRF families. The mutations were associated with lack of SCARB2 protein. Reanalysis of an existing Limp2 knockout mouse showed intracellular inclusions in cerebral and cerebellar cortex, and the kidneys showed subtle glomerular changes. This study highlights that recessive genes can be identified with a very small number of subjects. The ancestral lysosomal-membrane protein SCARB2/LIMP-2 is responsible for AMRF. The heterogeneous pathology in the kidney and brain suggests that SCARB2/Limp2 has pleiotropic effects that may be relevant to understanding the pathogenesis of other forms of glomerulosclerosis or collapse and myoclonic epilepsies.


Asunto(s)
Cromosomas Humanos Par 4/genética , Genes Recesivos , Glomerulonefritis/genética , Proteínas de Membrana de los Lisosomas/genética , Epilepsias Mioclónicas Progresivas/genética , Receptores Depuradores/genética , Animales , Corteza Cerebelosa/patología , Mapeo Cromosómico , Expresión Génica , Ligamiento Genético , Genotipo , Glomerulonefritis/patología , Humanos , Ratones , Ratones Noqueados , Epilepsias Mioclónicas Progresivas/patología , Análisis de Secuencia por Matrices de Oligonucleótidos
12.
JACC Basic Transl Sci ; 6(4): 365-380, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33997522

RESUMEN

Autophagy is a cellular degradation process that has been implicated in diverse disease processes. The authors provide evidence that FYCO1, a component of the autophagic machinery, is essential for adaptation to cardiac stress. Although the absence of FYCO1 does not affect basal autophagy in isolated cardiomyocytes, it abolishes induction of autophagy after glucose deprivation. Likewise, Fyco1-deficient mice subjected to starvation or pressure overload are unable to respond with induction of autophagy and develop impaired cardiac function. FYCO1 overexpression leads to induction of autophagy in isolated cardiomyocytes and transgenic mouse hearts, thereby rescuing cardiac dysfunction in response to biomechanical stress.

13.
Hum Mol Genet ; 17(22): 3437-45, 2008 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-18713755

RESUMEN

Despite the progress in the treatment of lysosomal storage disorders (LSDs) mainly by enzyme replacement therapy, only limited success was reported in targeting the appropriate lysosomal enzyme into the brain. This prevents efficient clearance of neuronal storage, which is present in many of these disorders including alpha-mannosidosis. Here we show that the neuropathology of a mouse model for alpha-mannosidosis can be efficiently treated using recombinant human alpha-mannosidase (rhLAMAN). After intravenous administration of different doses (25-500 U/kg), rhLAMAN was widely distributed among tissues, and immunohistochemistry revealed lysosomal delivery of the injected enzyme. Whereas low doses (25 U/kg) led to a significant clearance (<70%) in visceral tissues, higher doses were needed for a clear effect in central and peripheral nervous tissues. A distinct reduction (<50%) of brain storage required repeated high-dose injections (500 U/kg), whereas lower doses (250 U/kg) were sufficient for clearance of stored substrates in peripheral neurons of the trigeminal ganglion. Successful transfer across the blood-brain barrier was evident as the injected enzyme was found in hippocampal neurons, leading to a nearly complete disappearance of storage vacuoles. Importantly, the decrease in neuronal storage in the brain correlated with an improvement of the neuromotor disabilities found in untreated alpha-mannosidosis mice. Uptake of rhLAMAN seems to be independent of mannose-6-phosphate receptors, which is consistent with the low phosphorylation profile of the enzyme. These data suggest that high-dose injections of low phosphorylated enzymes might be an interesting option to efficiently treat LSDs with CNS involvement.


Asunto(s)
Ataxia/tratamiento farmacológico , Encéfalo/efectos de los fármacos , alfa-Manosidasa/uso terapéutico , alfa-Manosidosis/tratamiento farmacológico , Animales , Barrera Hematoencefálica , Encéfalo/metabolismo , Encéfalo/ultraestructura , Modelos Animales de Enfermedad , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Hipocampo/ultraestructura , Humanos , Riñón/efectos de los fármacos , Riñón/metabolismo , Riñón/ultraestructura , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/ultraestructura , Lisosomas/metabolismo , Ratones , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Receptor IGF Tipo 2/metabolismo , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/uso terapéutico , Bazo/efectos de los fármacos , Bazo/metabolismo , Bazo/ultraestructura , Ganglio del Trigémino/efectos de los fármacos , Ganglio del Trigémino/ultraestructura , Vacuolas/metabolismo , alfa-Manosidasa/administración & dosificación , alfa-Manosidasa/farmacocinética , alfa-Manosidasa/farmacología , alfa-Manosidosis/genética , alfa-Manosidosis/metabolismo , alfa-Manosidosis/patología
14.
Mol Ther ; 17(4): 600-6, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19174759

RESUMEN

Inherited deficiencies of lysosomal hydrolases cause lysosomal storage diseases (LSDs) that are characterized by a progressive multisystemic pathology and premature death. Repeated intravenous injection of the active counterpart of the deficient enzyme, a treatment strategy called enzyme replacement therapy (ERT), evolved as a clinical option for several LSDs without central nervous system (CNS) involvement. To assess the efficacy of long-term ERT in metachromatic leukodystrophy (MLD), an LSD with prevailing nervous system disease, we treated immunotolerant arylsulfatase A (ASA) knockout mice with 52 doses of either 4 or 50 mg/kg recombinant human ASA (rhASA). ERT was tolerated without side effects and improved disease manifestations in a dose-dependent manner. Dosing of 4 mg/kg diminished sulfatide storage in kidney and peripheral nervous system (PNS) but not the CNS, whereas treatment with 50 mg/kg was also effective in the CNS in reducing storage in brain and spinal cord by 34 and 45%, respectively. Histological analyses revealed regional differences in sulfatide clearance. While 70% less storage profiles were detectable, for example, in the hippocampal fimbria, the histopathology of the brain stem was unchanged. Both enzyme doses normalized the ataxic gait of ASA knockout mice, demonstrating prevention of nervous system dysfunctions that dominate early stages of MLD.


Asunto(s)
Ataxia/terapia , Sistema Nervioso Central/patología , Cerebrósido Sulfatasa/uso terapéutico , Modelos Animales de Enfermedad , Marcha , Leucodistrofia Metacromática/terapia , Animales , Ataxia/fisiopatología , Conducta Animal , Humanos , Ratones , Ratones Noqueados , Proteínas Recombinantes/uso terapéutico
15.
Cells ; 9(11)2020 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-33142804

RESUMEN

Dysbindin, a schizophrenia susceptibility marker and an essential constituent of BLOC-1 (biogenesis of lysosome-related organelles complex-1), has recently been associated with cardiomyocyte hypertrophy through the activation of Myozap-RhoA-mediated SRF signaling. We employed sandy mice (Dtnbp1_KO), which completely lack Dysbindin protein because of a spontaneous deletion of introns 5-7 of the Dtnbp1 gene, for pathophysiological characterization of the heart. Unlike in vitro, the loss-of-function of Dysbindin did not attenuate cardiac hypertrophy, either in response to transverse aortic constriction stress or upon phenylephrine treatment. Interestingly, however, the levels of hypertrophy-inducing interaction partner Myozap as well as the BLOC-1 partners of Dysbindin like Muted and Pallidin were dramatically reduced in Dtnbp1_KO mouse hearts. Taken together, our data suggest that Dysbindin's role in cardiomyocyte hypertrophy is redundant in vivo, yet essential to maintain the stability of its direct interaction partners like Myozap, Pallidin and Muted.


Asunto(s)
Cardiomegalia/genética , Cardiomegalia/metabolismo , Disbindina/genética , Disbindina/metabolismo , Proteínas Musculares/metabolismo , Miocitos Cardíacos/metabolismo , Animales , Citosol/metabolismo , Regulación de la Expresión Génica , Hipertrofia/fisiopatología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Biogénesis de Organelos , Unión Proteica , Esquizofrenia/genética , Esquizofrenia/metabolismo , Factor de Respuesta Sérica/metabolismo , Transducción de Señal , Proteínas de Transporte Vesicular/metabolismo , Proteína de Unión al GTP rhoA/metabolismo
16.
Cell Rep ; 30(10): 3506-3519.e6, 2020 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-32160553

RESUMEN

Genetic variations in TMEM106B, coding for a lysosomal membrane protein, affect frontotemporal lobar degeneration (FTLD) in GRN- (coding for progranulin) and C9orf72-expansion carriers and might play a role in aging. To determine the physiological function of TMEM106B, we generated TMEM106B-deficient mice. These mice develop proximal axonal swellings caused by drastically enlarged LAMP1-positive vacuoles, increased retrograde axonal transport of lysosomes, and accumulation of lipofuscin and autophagosomes. Giant vacuoles specifically accumulate at the distal end and within the axon initial segment, but not in peripheral nerves or at axon terminals, resulting in an impaired facial-nerve-dependent motor performance. These data implicate TMEM106B in mediating the axonal transport of LAMP1-positive organelles in motoneurons and axonal sorting at the initial segment. Our data provide mechanistic insight into how TMEM106B affects lysosomal proteolysis and degradative capacity in neurons.


Asunto(s)
Segmento Inicial del Axón/metabolismo , Degeneración Lobar Frontotemporal/genética , Predisposición Genética a la Enfermedad , Lisosomas/metabolismo , Proteínas de la Membrana/genética , Neuronas Motoras/metabolismo , Proteínas del Tejido Nervioso/genética , Animales , Autofagosomas/metabolismo , Autofagosomas/ultraestructura , Segmento Inicial del Axón/ultraestructura , Transporte Axonal , Tronco Encefálico/patología , Núcleo Celular/metabolismo , Nervio Facial/patología , Lisosomas/ultraestructura , Proteínas de la Membrana/deficiencia , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas Motoras/ultraestructura , Músculos/inervación , Proteínas del Tejido Nervioso/deficiencia , Factores de Riesgo
17.
Autophagy ; 16(5): 811-825, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31282275

RESUMEN

CTSD (cathepsin D) is one of the major lysosomal proteases indispensable for the maintenance of cellular proteostasis by turning over substrates of endocytosis, phagocytosis and autophagy. Consequently, CTSD deficiency leads to a strong impairment of the lysosomal-autophagy machinery. In mice and humans CTSD dysfunction underlies the congenital variant (CLN10) of neuronal ceroid lipofuscinosis (NCL). NCLs are distinct lysosomal storage disorders (LSDs) sharing various hallmarks, namely accumulation of protein aggregates and ceroid lipofuscin leading to neurodegeneration and blindness. The most established and clinically approved approach to treat LSDs is enzyme replacement therapy (ERT) aiming to replace the defective hydrolase with an exogenously applied recombinant protein. Here we reveal that recombinant human pro-CTSD produced in a mammalian expression system can be efficiently taken up by a variety of cell models, is correctly targeted to lysosomes and processed to the active mature form of the protease. In proof-of-principle experiments we provide evidence that recombinant human CTSD (rhCTSD) can improve the biochemical phenotype of CTSD-deficient hippocampal slice cultures in vitro and retinal cells in vivo. Furthermore, we demonstrate that dosing of rhCTSD in the murine CLN10 model leads to a correction of lysosomal hypertrophy, storage accumulation and impaired autophagic flux in the viscera and central nervous system (CNS). We establish that direct delivery of the recombinant protease to the CNS is required for improvement of neuropathology and lifespan extension. Together these data support the continuation of the pre-clinical studies for the application of rhCTSD in the treatment of NCL.Abbreviations: AIF1/IBA1: allograft inflammatory factor 1; BBB: blood brain barrier; CNS: central nervous system; CTSB: cathepsin B; CTSD: cathepsin D; CTSL: cathepsin L; ERT: enzyme replacement therapy; GFAP: glial fibrillary acidic protein; INL: inner nuclear layer; LAMP1: lysosomal-associated membrane protein 1; LAMP2: lysosomal-associated membrane protein 2; MAP1LC3/LC3: microtubule-associated protein 1 light chain 3; LDL: low-density lipoprotein; LRP1: low density lipoprotein receptor-related protein 1; LSD: lysosomal storage disorder; MEFs: mouse embryonic fibroblasts; M6P: mannose 6-phosphate; mCTSD: mature CTSD; NCL: neuronal ceroid lipofuscinosis; ONL: outer nuclear layer; PB: phosphate buffer; proCTSD: pro-cathepsin D; LRPAP1: low density lipoprotein receptor-related protein associated protein 1; rhCTSD: human recombinant CTSD; SAPC: saposin C; SAPD: saposin D; ATP5G1: ATP synthase, H+ transporting, mitochondrial F0 complex, subunit C1 (subunit 9); SQSTM1/p62: sequestosome 1; TPP1: tripeptidyl peptidase I.


Asunto(s)
Autofagia/efectos de los fármacos , Catepsina D/uso terapéutico , Terapia de Reemplazo Enzimático , Lipofuscinosis Ceroideas Neuronales/tratamiento farmacológico , Lipofuscinosis Ceroideas Neuronales/metabolismo , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Catepsina D/metabolismo , Modelos Animales de Enfermedad , Terapia de Reemplazo Enzimático/métodos , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Ratones Noqueados , Tripeptidil Peptidasa 1
18.
Elife ; 82019 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-31661432

RESUMEN

Lysosomes are major sites for intracellular, acidic hydrolase-mediated proteolysis and cellular degradation. The export of low-molecular-weight catabolic end-products is facilitated by polytopic transmembrane proteins mediating secondary active or passive transport. A number of these lysosomal transporters, however, remain enigmatic. We present a detailed analysis of MFSD1, a hitherto uncharacterized lysosomal family member of the major facilitator superfamily. MFSD1 is not N-glycosylated. It contains a dileucine-based sorting motif needed for its transport to lysosomes. Mfsd1 knockout mice develop splenomegaly and severe liver disease. Proteomics of isolated lysosomes from Mfsd1 knockout mice revealed GLMP as a critical accessory subunit for MFSD1. MFSD1 and GLMP physically interact. GLMP is essential for the maintenance of normal levels of MFSD1 in lysosomes and vice versa. Glmp knockout mice mimic the phenotype of Mfsd1 knockout mice. Our data reveal a tightly linked MFSD1/GLMP lysosomal membrane protein transporter complex.


Asunto(s)
Hígado/fisiología , Lisosomas/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Animales , Homeostasis , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Proteínas de Transporte de Membrana/deficiencia , Proteínas de Transporte de Membrana/genética , Ratones Noqueados , Unión Proteica
19.
Nat Commun ; 10(1): 3521, 2019 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-31387993

RESUMEN

The intracellular transport of cholesterol is subject to tight regulation. The structure of the lysosomal integral membrane protein type 2 (LIMP-2, also known as SCARB2) reveals a large cavity that traverses the molecule and resembles the cavity in SR-B1 that mediates lipid transfer. The detection of cholesterol within the LIMP-2 structure and the formation of cholesterol-like inclusions in LIMP-2 knockout mice suggested the possibility that LIMP2 transports cholesterol in lysosomes. We present results of molecular modeling, crosslinking studies, microscale thermophoresis and cell-based assays that support a role of LIMP-2 in cholesterol transport. We show that the cavity in the luminal domain of LIMP-2 can bind and deliver exogenous cholesterol to the lysosomal membrane and later to lipid droplets. Depletion of LIMP-2 alters SREBP-2-mediated cholesterol regulation, as well as LDL-receptor levels. Our data indicate that LIMP-2 operates in parallel with Niemann Pick (NPC)-proteins, mediating a slower mode of lysosomal cholesterol export.


Asunto(s)
Antígenos CD36/metabolismo , LDL-Colesterol/metabolismo , Proteínas de Membrana de los Lisosomas/metabolismo , Lisosomas/metabolismo , Receptores Depuradores/metabolismo , Animales , Antígenos CD36/genética , Células CHO , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Cricetulus , Fibroblastos , Técnicas de Inactivación de Genes , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intracelular , Gotas Lipídicas/metabolismo , Proteínas de Membrana de los Lisosomas/genética , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ratones , Proteína Niemann-Pick C1 , Dominios Proteicos , ARN Interferente Pequeño/metabolismo , Receptores Depuradores/genética
20.
J Neurosci ; 27(34): 9009-21, 2007 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-17715338

RESUMEN

Metachromatic leukodystrophy is a lysosomal storage disorder caused by deficiency in the sulfolipid degrading enzyme arylsulfatase A (ASA). In the absence of a functional ASA gene, 3-O-sulfogalactosylceramide (sulfatide; SGalCer) and other sulfolipids accumulate. The storage is associated with progressive demyelination and various finally lethal neurological symptoms. Lipid storage, however, is not restricted to myelin-producing cells but also occurs in neurons. It is unclear whether neuronal storage contributes to symptoms of the patients. Therefore, we have generated transgenic ASA-deficient [ASA(-/-)] mice overexpressing the sulfatide synthesizing enzymes UDP-galactose:ceramide galactosyltransferase (CGT) and cerebroside sulfotransferase (CST) in neurons to provoke neuronal lipid storage. CGT-transgenic ASA(-/-) [CGT/ASA(-/-)] mice showed an accumulation of C18:0 fatty acid-containing SGalCer in the brain. Histochemically, an increase in sulfolipid storage could be detected in central and peripheral neurons of both CGT/ASA(-/-) and CST/ASA(-/-) mice compared with ASA(-/-) mice. CGT/ASA(-/-) mice developed severe neuromotor coordination deficits and weakness of hindlimbs and forelimbs. Light and electron microscopic analyses demonstrated nerve fiber degeneration in the spinal cord of CGT/ASA(-/-) mice. CGT/ASA(-/-) and, to a lesser extent, young ASA(-/-) mice exhibited cortical hyperexcitability, with recurrent spontaneous cortical EEG discharges lasting 5-15 s. These observations suggest that SGalCer accumulation in neurons contributes to disease phenotype.


Asunto(s)
Corteza Cerebral/fisiopatología , Cerebrósido Sulfatasa/metabolismo , Leucodistrofia Metacromática , Degeneración Nerviosa/etiología , Neuronas/enzimología , Sulfoglicoesfingolípidos/metabolismo , Análisis de Varianza , Animales , Conducta Animal , Corteza Cerebral/patología , Cerebrósido Sulfatasa/deficiencia , Modelos Animales de Enfermedad , Electroencefalografía/métodos , Hibridación in Situ/métodos , Leucodistrofia Metacromática/complicaciones , Leucodistrofia Metacromática/metabolismo , Leucodistrofia Metacromática/patología , Lípidos/análisis , Ratones , Ratones Transgénicos , Microscopía Electrónica de Transmisión , Destreza Motora/fisiología , N-Acilesfingosina Galactosiltransferasa/deficiencia , Degeneración Nerviosa/genética , Neuronas/ultraestructura , Ratas , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Médula Espinal/patología , Sulfotransferasas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA