Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Kidney Int ; 104(5): 995-1007, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37598857

RESUMEN

Autosomal dominant polycystic kidney disease (ADPKD) resulting from pathogenic variants in PKD1 and PKD2 is the most common form of PKD, but other genetic causes tied to primary cilia function have been identified. Biallelic pathogenic variants in the serine/threonine kinase NEK8 cause a syndromic ciliopathy with extra-kidney manifestations. Here we identify NEK8 as a disease gene for ADPKD in 12 families. Clinical evaluation was combined with functional studies using fibroblasts and tubuloids from affected individuals. Nek8 knockout mouse kidney epithelial (IMCD3) cells transfected with wild type or variant NEK8 were further used to study ciliogenesis, ciliary trafficking, kinase function, and DNA damage responses. Twenty-one affected monoallelic individuals uniformly exhibited cystic kidney disease (mostly neonatal) without consistent extra-kidney manifestations. Recurrent de novo mutations of the NEK8 missense variant p.Arg45Trp, including mosaicism, were seen in ten families. Missense variants elsewhere within the kinase domain (p.Ile150Met and p.Lys157Gln) were also identified. Functional studies demonstrated normal localization of the NEK8 protein to the proximal cilium and no consistent cilia formation defects in patient-derived cells. NEK8-wild type protein and all variant forms of the protein expressed in Nek8 knockout IMCD3 cells were localized to cilia and supported ciliogenesis. However, Nek8 knockout IMCD3 cells expressing NEK8-p.Arg45Trp and NEK8-p.Lys157Gln showed significantly decreased polycystin-2 but normal ANKS6 localization in cilia. Moreover, p.Arg45Trp NEK8 exhibited reduced kinase activity in vitro. In patient derived tubuloids and IMCD3 cells expressing NEK8-p.Arg45Trp, DNA damage signaling was increased compared to healthy passage-matched controls. Thus, we propose a dominant-negative effect for specific heterozygous missense variants in the NEK8 kinase domain as a new cause of PKD.


Asunto(s)
Enfermedades Renales Poliquísticas , Riñón Poliquístico Autosómico Dominante , Animales , Humanos , Recién Nacido , Ratones , Proteínas Portadoras/metabolismo , Cilios/patología , Riñón/metabolismo , Mutación , Quinasas Relacionadas con NIMA/genética , Quinasas Relacionadas con NIMA/metabolismo , Enfermedades Renales Poliquísticas/genética , Riñón Poliquístico Autosómico Dominante/patología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Serina/genética , Serina/metabolismo , Canales Catiónicos TRPP/genética , Canales Catiónicos TRPP/metabolismo
2.
JCI Insight ; 7(13)2022 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-35801587

RESUMEN

Polyamine dysregulation plays key roles in a broad range of human diseases from cancer to neurodegeneration. Snyder-Robinson syndrome (SRS) is the first known genetic disorder of the polyamine pathway, caused by X-linked recessive loss-of-function mutations in spermine synthase. In the Drosophila SRS model, altered spermidine/spermine balance has been associated with increased generation of ROS and aldehydes, consistent with elevated spermidine catabolism. These toxic byproducts cause mitochondrial and lysosomal dysfunction, which are also observed in cells from SRS patients. No efficient therapy is available. We explored the biochemical mechanism and discovered acetyl-CoA reduction and altered protein acetylation as potentially novel pathomechanisms of SRS. We repurposed the FDA-approved drug phenylbutyrate (PBA) to treat SRS using an in vivo Drosophila model and patient fibroblast cell models. PBA treatment significantly restored the function of mitochondria and autolysosomes and extended life span in vivo in the Drosophila SRS model. Treating fibroblasts of patients with SRS with PBA ameliorated autolysosome dysfunction. We further explored the mechanism of drug action and found that PBA downregulates the first and rate-limiting spermidine catabolic enzyme spermidine/spermine N1-acetyltransferase 1 (SAT1), reduces the production of toxic metabolites, and inhibits the reduction of the substrate acetyl-CoA. Taken together, we revealed PBA as a potential modulator of SAT1 and acetyl-CoA levels and propose PBA as a therapy for SRS and potentially other polyamine dysregulation-related diseases.


Asunto(s)
Poliaminas , Espermidina , Acetilcoenzima A/metabolismo , Acetilesterasa , Acetiltransferasas/genética , Acetiltransferasas/metabolismo , Animales , Drosophila/metabolismo , Discapacidad Intelectual Ligada al Cromosoma X , Fenilbutiratos/farmacología , Poliaminas/metabolismo , Espermidina/metabolismo , Espermina/metabolismo
3.
Brain Commun ; 4(3): fcac102, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35602653

RESUMEN

Altered brain iron homeostasis can contribute to neurodegeneration by interfering with the delivery of the iron needed to support key cellular processes, including mitochondrial respiration, synthesis of myelin and essential neurotransmitters. Intracellular iron homeostasis in mammals is maintained by two homologous ubiquitously expressed iron-responsive element-binding proteins (IRP1 and IRP2). Using exome sequencing, two patients with severe neurodegenerative disease and bi-allelic mutations in the gene IREB2 were first identified and clinically characterized in 2019. Here, we report the case of a 7-year-old male patient with compound heterozygous missense variants in IREB2, whose neurological features resembled those of the two previously reported IRP2-deficient patients, including a profound global neurodevelopmental delay and dystonia. Biochemical characterization of a lymphoblast cell line derived from the patient revealed functional iron deficiency, altered post-transcriptional regulation of iron metabolism genes and mitochondrial dysfunction. The iron metabolism abnormalities of the patient cell line were reversed by lentiviral-mediated restoration of IREB2 expression. These results, in addition to confirming the essential role of IRP2 in the regulation of iron metabolism in humans, expand the scope of the known IRP2-related neurodegenerative disorders and underscore that IREB2 pathological variants may impact the iron-responsive element-binding activity of IRP2 with varying degrees of severity. The three severely affected patients identified so far all suffered from complete loss of function of IRP2, raising the possibility that individuals with significant but incomplete loss of IRP2 function may develop less severe forms of the disease, analogous to other human conditions that present with a wide range of phenotypic manifestations.

4.
Am J Hum Genet ; 108(6): 1040-1052, 2021 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-33964207

RESUMEN

SLC37A4 encodes an endoplasmic reticulum (ER)-localized multitransmembrane protein required for transporting glucose-6-phosphate (Glc-6P) into the ER. Once transported into the ER, Glc-6P is subsequently hydrolyzed by tissue-specific phosphatases to glucose and inorganic phosphate during times of glucose depletion. Pathogenic variants in SLC37A4 cause an established recessive disorder known as glycogen storage disorder 1b characterized by liver and kidney dysfunction with neutropenia. We report seven individuals who presented with liver dysfunction multifactorial coagulation deficiency and cardiac issues and were heterozygous for the same variant, c.1267C>T (p.Arg423∗), in SLC37A4; the affected individuals were from four unrelated families. Serum samples from affected individuals showed profound accumulation of both high mannose and hybrid type N-glycans, while N-glycans in fibroblasts and undifferentiated iPSC were normal. Due to the liver-specific nature of this disorder, we generated a CRISPR base-edited hepatoma cell line harboring the c.1267C>T (p.Arg423∗) variant. These cells replicated the secreted abnormalities seen in serum N-glycosylation, and a portion of the mutant protein appears to relocate to a distinct, non-Golgi compartment, possibly ER exit sites. These cells also show a gene dosage-dependent alteration in the Golgi morphology and reduced intraluminal pH that may account for the altered glycosylation. In summary, we identify a recurrent mutation in SLC37A4 that causes a dominantly inherited congenital disorder of glycosylation characterized by coagulopathy and liver dysfunction with abnormal serum N-glycans.


Asunto(s)
Antiportadores/genética , Trastornos Congénitos de Glicosilación/etiología , Retículo Endoplásmico/patología , Hepatopatías/complicaciones , Proteínas de Transporte de Monosacáridos/genética , Mutación , Adulto , Niño , Preescolar , Trastornos Congénitos de Glicosilación/patología , Retículo Endoplásmico/genética , Retículo Endoplásmico/metabolismo , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Genes Dominantes , Glicosilación , Humanos , Lactante , Recién Nacido , Masculino , Linaje
5.
Sci Adv ; 7(3)2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33523898

RESUMEN

Asparagine (N)-linked glycosylation is required for endoplasmic reticulum (ER) homeostasis, but how this co- and posttranslational modification is maintained during ER stress is unknown. Here, we introduce a fluorescence-based strategy to detect aberrant N-glycosylation in individual cells and identify a regulatory role for the heterotetrameric translocon-associated protein (TRAP) complex. Unexpectedly, cells with knockout of SSR3 or SSR4 subunits restore N-glycosylation over time concurrent with a diminished ER stress transcriptional signature. Activation of ER stress or silencing of the ER chaperone BiP exacerbates or rescues the glycosylation defects, respectively, indicating that SSR3 and SSR4 enable N-glycosylation during ER stress. Protein levels of the SSR3 subunit are ER stress and UBE2J1 dependent, revealing a mechanism that coordinates upstream N-glycosylation proficiency with downstream ER-associated degradation and proteostasis. The fidelity of N-glycosylation is not static in both nontransformed and tumor cells, and the TRAP complex regulates ER glycoprotein quality control under conditions of stress.

6.
Cells ; 9(6)2020 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-32517081

RESUMEN

The glycoprotein disorders are a group of lysosomal storage diseases (α-mannosidosis, aspartylglucosaminuria, ß-mannosidosis, fucosidosis, galactosialidosis, sialidosis, mucolipidosis II, mucolipidosis III, and Schindler Disease) characterized by specific lysosomal enzyme defects and resultant buildup of undegraded glycoprotein substrates. This buildup causes a multitude of abnormalities in patients including skeletal dysplasia, inflammation, ocular abnormalities, liver and spleen enlargement, myoclonus, ataxia, psychomotor delay, and mild to severe neurodegeneration. Pharmacological treatment options exist through enzyme replacement therapy (ERT) for a few, but therapies for this group of disorders is largely lacking. Hematopoietic cell transplant (HCT) has been explored as a potential therapeutic option for many of these disorders, as HCT introduces functional enzyme-producing cells into the bone marrow and blood along with the engraftment of healthy donor cells in the central nervous system (presumably as brain macrophages or a type of microglial cell). The outcome of HCT varies widely by disease type. We report our institutional experience with HCT as well as a review of the literature to better understand HCT and outcomes for the glycoprotein disorders.


Asunto(s)
Glicoproteínas/metabolismo , Trasplante de Células Madre Hematopoyéticas , Enfermedades por Almacenamiento Lisosomal/terapia , Animales , Terapia de Reemplazo Enzimático , Humanos
7.
JCI Insight ; 3(14)2018 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-30046013

RESUMEN

Site-1 protease (S1P), encoded by MBTPS1, is a serine protease in the Golgi. S1P regulates lipogenesis, endoplasmic reticulum (ER) function, and lysosome biogenesis in mice and in cultured cells. However, how S1P differentially regulates these diverse functions in humans has been unclear. In addition, no human disease with S1P deficiency has been identified. Here, we report a pediatric patient with an amorphic and a severely hypomorphic mutation in MBTPS1. The unique combination of these mutations results in a frequency of functional MBTPS1 transcripts of approximately 1%, a finding that is associated with skeletal dysplasia and elevated blood lysosomal enzymes. We found that the residually expressed S1P is sufficient for lipid homeostasis but not for ER and lysosomal functions, especially in chondrocytes. The defective S1P function specifically impairs activation of the ER stress transducer BBF2H7, leading to ER retention of collagen in chondrocytes. S1P deficiency also causes abnormal secretion of lysosomal enzymes due to partial impairment of mannose-6-phosphate-dependent delivery to lysosomes. Collectively, these abnormalities lead to apoptosis of chondrocytes and lysosomal enzyme-mediated degradation of the bone matrix. Correction of an MBTPS1 variant or reduction of ER stress mitigated collagen-trafficking defects. These results define a new congenital human skeletal disorder and, more importantly, reveal that S1P is particularly required for skeletal development in humans. Our findings may also lead to new therapies for other genetic skeletal diseases, as ER dysfunction is common in these disorders.


Asunto(s)
Enfermedades del Desarrollo Óseo/genética , Enfermedades del Desarrollo Óseo/metabolismo , Proproteína Convertasas/genética , Proproteína Convertasas/metabolismo , Transporte de Proteínas , Serina Endopeptidasas/genética , Serina Endopeptidasas/metabolismo , Apoptosis , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Enfermedades del Desarrollo Óseo/fisiopatología , Técnicas de Cultivo de Célula , Preescolar , Condrocitos/metabolismo , Colágeno/metabolismo , Retículo Endoplásmico/metabolismo , Femenino , Técnicas de Silenciamiento del Gen , Enfermedades Genéticas Congénitas , Aparato de Golgi/metabolismo , Homeostasis , Humanos , Lipogénesis , Lisosomas/metabolismo , Manosafosfatos , Mutación
8.
Cell Rep ; 22(11): 2964-2977, 2018 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-29539424

RESUMEN

Cysteine cathepsins play roles during development and disease beyond their function in lysosomal protein turnover. Here, we leverage a fluorescent activity-based probe (ABP), BMV109, to track cysteine cathepsins in normal and diseased zebrafish embryos. Using this probe in a model of mucolipidosis II, we show that loss of carbohydrate-dependent lysosomal sorting alters the activity of several cathepsin proteases. The data support a pathogenic mechanism where TGF-ß signals enhance the proteolytic processing of pro-Ctsk by modulating the expression of chondroitin 4-sulfate (C4-S). In MLII, elevated C4-S corresponds with TGF-ß-mediated increases in chst11 expression. Inhibiting chst11 impairs the proteolytic activation of Ctsk and alleviates the MLII phenotypes. These findings uncover a regulatory loop between TGF-ß signaling and Ctsk activation that is altered in the context of lysosomal disease. This work highlights the power of ABPs to identify mechanisms underlying pathogenic development in living animals.


Asunto(s)
Catepsinas/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Modelos Animales de Enfermedad , Pez Cebra
9.
J Biol Chem ; 292(36): 15094-15104, 2017 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-28724630

RESUMEN

Acid hydrolases utilize a carbohydrate-dependent mechanism for lysosomal targeting. These hydrolases acquire a mannose 6-phosphate tag by the action of the GlcNAc-1-phosphotransferase enzyme, allowing them to bind receptors and traffic to endosomes. Loss of GlcNAc-1-phosphotransferase results in hydrolase hypersecretion and profound lysosomal storage. Little, however, is known about how these cellular phenotypes affect the trafficking, activity, and localization of surface glycoproteins. To address this question, we profiled the abundance of surface glycoproteins in WT and CRISPR-mediated GNPTAB-/- HeLa cells and identified changes in numerous glycoproteins, including the uptake receptor LRP1 and multiple receptor tyrosine kinases. Decreased cell surface LRP1 in GNPTAB-/- cells corresponded with a reduction in its steady-state level and less amyloid-ß-40 (Aß40) peptide uptake. GNPTAB-/- cells displayed elevated activation of several kinases including Met receptor. We found increased Met phosphorylation within both the kinase and the docking domains and observed that lower concentrations of pervanadate were needed to cause an increase in phospho-Met in GNPTAB-/- cells. Together, these data suggested a decrease in the activity of the receptor and non-receptor protein-tyrosine phosphatases that down-regulate Met phosphorylation. GNPTAB-/- cells exhibited elevated levels of reactive oxygen species, known to inactivate cell surface and cytosolic phosphatases by oxidation of active site cysteine residues. Consistent with this mode of action, peroxide treatment of parental HeLa cells elevated phospho-Met levels whereas antioxidant treatment of GNPTAB-/- cells reduced phospho-Met levels. Collectively, these findings identify new mechanisms whereby impaired lysosomal targeting can impact the activity and recycling of receptors.


Asunto(s)
Carbohidratos , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Lisosomas/metabolismo , Proteínas Proto-Oncogénicas c-met/metabolismo , Transferasas (Grupos de Otros Fosfatos Sustitutos)/metabolismo , Células HeLa , Humanos , Fosforilación , Proteínas Proto-Oncogénicas c-met/química , Transferasas (Grupos de Otros Fosfatos Sustitutos)/deficiencia , Transferasas (Grupos de Otros Fosfatos Sustitutos)/genética , Células Tumorales Cultivadas
10.
J Biol Chem ; 291(8): 3982-9, 2016 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-26733198

RESUMEN

Selective exo-enzymatic labeling (or SEEL) uses recombinant glycosyltransferases and nucleotide-sugar analogues to allow efficient labeling of cell surface glycans. SEEL can circumvent many of the possible issues associated with metabolic labeling, including low incorporation of sugar precursors, and allows for sugars to be added selectively to different types of glycans by virtue of the inherent specificity of the glycosyltransferases. Here we compare the labeling of sialoglycoproteins in undifferentiated and differentiated human erythroleukemia cells (HEL) using SEEL using the sialyltransferases ST6Gal1 and ST3Gal1, which label N- and O-glycans, respectively. Our results show that the profile of glycoproteins detected varies between undifferentiated HEL cells and those differentiated to megakaryocytes, with a shift to more N-linked sialoglycoproteins in the differentiated cells. The efficiency of SEEL for both sialyltransferases in HEL cells was greatly increased with prior neuraminidase treatment highlighting the necessity for the presence of available acceptors with this labeling method. Following metabolic labeling or SEEL, tagged glycoproteins were enriched by immunoprecipitation and identified using mass spectrometry. The proteomic findings demonstrated that the detection of many glycoproteins is markedly improved by SEEL labeling, and that unique glycoproteins can be identified using either ST6Gal1 or ST3Gal1. Furthermore, this analysis enabled the identification of increased surface expression of several sialylated cell adhesion molecules, including the known megakaryocytic markers integrinß3 and CD44, upon differentiation of HEL cells to adherent megakaryocytes.


Asunto(s)
Marcaje Isotópico/métodos , Megacariocitos/metabolismo , Sialoglicoproteínas/biosíntesis , Animales , Antígenos CD/metabolismo , Células CHO , Cricetinae , Cricetulus , Humanos , Receptores de Hialuranos/biosíntesis , Integrina beta3/biosíntesis , Megacariocitos/citología , Sialiltransferasas/metabolismo , beta-Galactosida alfa-2,3-Sialiltransferasa
11.
Mol Ther ; 24(2): 206-216, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26447927

RESUMEN

Mucopolysaccharidosis VII (MPS VII) is a lysosomal storage disease arising from mutations in ß-d-glucuronidase (GUSB), which results in glycosaminoglycan (GAG) accumulation and a variety of clinical manifestations including neurological disease. Herein, MPS VII dogs were injected intravenously (i.v.) and/or intrathecally (i.t.) via the cisterna magna with AAV9 or AAVrh10 vectors carrying the canine GUSB cDNA. Although i.v. injection alone at 3 days of age resulted in normal cerebrospinal fluid (CSF) GUSB activity, brain tissue homogenates had only ~1 to 6% normal GUSB activity and continued to have elevated GAG storage. In contrast, i.t. injection at 3 weeks of age resulted in CSF GUSB activity 44-fold normal while brain tissue homogenates had >100% normal GUSB activity and reduced GAGs compared with untreated dogs. Markers for secondary storage and inflammation were eliminated in i.t.-treated dogs and reduced in i.v.-treated dogs compared with untreated dogs. Given that i.t.-treated dogs expressed higher levels of GUSB in the CNS tissues compared to those treated i.v., we conclude that i.t. injection of AAV9 or AAVrh10 vectors is more effective than i.v. injection alone in the large animal model of MPS VII.


Asunto(s)
Enfermedades del Sistema Nervioso Central/terapia , Terapia Genética/métodos , Glucuronidasa/genética , Mucopolisacaridosis VII/terapia , Animales , Animales Recién Nacidos , Enfermedades del Sistema Nervioso Central/genética , Enfermedades del Sistema Nervioso Central/metabolismo , Dependovirus/genética , Modelos Animales de Enfermedad , Perros , Vectores Genéticos/administración & dosificación , Glucuronidasa/líquido cefalorraquídeo , Glicosaminoglicanos/metabolismo , Inyecciones Intravenosas , Inyecciones Espinales , Masculino , Mucopolisacaridosis VII/complicaciones , Mucopolisacaridosis VII/genética , Mucopolisacaridosis VII/metabolismo
12.
Glycobiology ; 25(6): 669-82, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25609749

RESUMEN

The congenital disorders of glycosylation (CDG), a group of inherited diseases characterized by aberrant glycosylation, encompass a wide range of defects, including glycosyltransferases, glycosidases, nucleotide-sugar transporters as well as proteins involved in maintaining Golgi architecture, pH and vesicular trafficking. Mutations in a previously undescribed protein, TMEM165, were recently shown to cause a new form of CDG, termed TMEM165-CDG. TMEM165-CDG patients exhibit cartilage and bone dysplasia and altered glycosylation of serum glycoproteins. We utilized a morpholino knockdown strategy in zebrafish to investigate the physiologic and pathogenic functions of TMEM165. Inhibition of tmem165 expression in developing zebrafish embryos caused craniofacial abnormalities, largely attributable to fewer chondrocytes. Decreased expression of several markers of cartilage and bone development suggests that Tmem165 deficiency alters both chondrocyte and osteoblast differentiation. Glycomic analysis of tmem165 morphants also revealed altered initiation, processing and extension of N-glycans, paralleling some of the glycosylation changes noted in human patients. Collectively, these findings highlight the utility of zebrafish to elucidate pathogenic mechanisms associated with glycosylation disorders and suggest that the cartilage and bone dysplasia manifested in TMEM165-CDG patients may stem from abnormal development of chondrocytes and osteoblasts.


Asunto(s)
Cartílago/metabolismo , Cartílago/patología , Trastornos Congénitos de Glicosilación/genética , Trastornos Congénitos de Glicosilación/metabolismo , Modelos Animales de Enfermedad , Proteínas de la Membrana/deficiencia , Pez Cebra/metabolismo , Animales , Antiportadores , Cartílago/crecimiento & desarrollo , Proteínas de Transporte de Catión , Glicosilación , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Fenotipo
13.
J Biol Chem ; 290(5): 3045-56, 2015 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-25505245

RESUMEN

UDP-GlcNAc:lysosomal enzyme GlcNAc-1-phosphotransferase tags newly synthesized lysosomal enzymes with mannose 6-phosphate recognition markers, which are required for their targeting to the endolysosomal system. GNPTAB encodes the α and ß subunits of GlcNAc-1-phosphotransferase, and mutations in this gene cause the lysosomal storage disorders mucolipidosis II and III αß. Prior investigation of missense mutations in GNPTAB uncovered amino acids in the N-terminal region and within the DMAP domain involved in Golgi retention of GlcNAc-1-phosphotransferase and its ability to specifically recognize lysosomal hydrolases, respectively. Here, we undertook a comprehensive analysis of the remaining missense mutations in GNPTAB reported in mucolipidosis II and III αß patients using cell- and zebrafish-based approaches. We show that the Stealth domain harbors the catalytic site, as some mutations in these regions greatly impaired the activity of the enzyme without affecting its Golgi localization and proteolytic processing. We also demonstrate a role for the Notch repeat 1 in lysosomal hydrolase recognition, as missense mutations in conserved cysteine residues in this domain do not affect the catalytic activity but impair mannose phosphorylation of certain lysosomal hydrolases. Rescue experiments using mRNA bearing Notch repeat 1 mutations in GNPTAB-deficient zebrafish revealed selective effects on hydrolase recognition that differ from the DMAP mutation. Finally, the mutant R587P, located in the spacer between Notch 2 and DMAP, was partially rescued by overexpression of the γ subunit, suggesting a role for this region in γ subunit binding. These studies provide new insight into the functions of the different domains of the α and ß subunits.


Asunto(s)
Lisosomas/metabolismo , Mucolipidosis/enzimología , Mucolipidosis/genética , Mutación Missense/genética , Transferasas (Grupos de Otros Fosfatos Sustitutos)/genética , Transferasas (Grupos de Otros Fosfatos Sustitutos)/metabolismo , Animales , Humanos , Mucolipidosis/metabolismo , Pez Cebra
14.
Hum Mol Genet ; 23(2): 418-33, 2014 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-24026681

RESUMEN

'Salt & Pepper' syndrome is an autosomal recessive condition characterized by severe intellectual disability, epilepsy, scoliosis, choreoathetosis, dysmorphic facial features and altered dermal pigmentation. High-density SNP array analysis performed on siblings first described with this syndrome detected four shared regions of loss of heterozygosity (LOH). Whole-exome sequencing narrowed the candidate region to chromosome 2p11.2. Sanger sequencing confirmed a homozygous c.994G>A transition (p.E332K) in the ST3GAL5 gene, which encodes for a sialyltransferase also known as GM3 synthase. A different homozygous mutation of this gene has been previously associated with infantile-onset epilepsy syndromes in two other cohorts. The ST3GAL5 enzyme synthesizes ganglioside GM3, a glycosophingolipid enriched in neural tissue, by adding sialic acid to lactosylceramide. Unlike disorders of glycosphingolipid (GSL) degradation, very little is known regarding the molecular and pathophysiologic consequences of altered GSL biosynthesis. Glycolipid analysis confirmed a complete lack of GM3 ganglioside in patient fibroblasts, while microarray analysis of glycosyltransferase mRNAs detected modestly increased expression of ST3GAL5 and greater changes in transcripts encoding enzymes that lie downstream of ST3GAL5 and in other GSL biosynthetic pathways. Comprehensive glycomic analysis of N-linked, O-linked and GSL glycans revealed collateral alterations in response to loss of complex gangliosides in patient fibroblasts and in zebrafish embryos injected with antisense morpholinos that targeted zebrafish st3gal5 expression. Morphant zebrafish embryos also exhibited increased apoptotic cell death in multiple brain regions, emphasizing the importance of GSL expression in normal neural development and function.


Asunto(s)
Gangliósido G(M3)/biosíntesis , Glucolípidos/metabolismo , Síndromes Neurocutáneos/genética , Sialiltransferasas/genética , Secuencia de Aminoácidos , Animales , Apoptosis , Cromosomas Humanos Par 2 , Secuencia Conservada , Embrión no Mamífero/metabolismo , Exoma , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Variación Genética , Glicoproteínas/metabolismo , Glicosilación , Humanos , Masculino , Datos de Secuencia Molecular , Síndromes Neurocutáneos/metabolismo , Neuronas/metabolismo , Linaje , Polimorfismo de Nucleótido Simple , Sialiltransferasas/química , Sialiltransferasas/metabolismo , Pez Cebra/embriología
15.
Glycoconj J ; 30(1): 33-40, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22638861

RESUMEN

Zebrafish (Danio rerio) remains a versatile model organism for the investigation of early development and organogenesis, and has emerged as a valuable platform for drug discovery and toxicity evaluation [1-6]. Harnessing the genetic power and experimental accessibility of this system, three decades of research have identified key genes and pathways that control the development of multiple organ systems and tissues, including the heart, kidney, and craniofacial cartilage, as well as the hematopoietic, vascular, and central and peripheral nervous systems [7-31]. In addition to their application in large mutagenic screens, zebrafish has been used to model a variety of diseases such as diabetes, polycystic kidney disease, muscular dystrophy and cancer [32-36]. As this work continues to intersect with cellular pathways and processes such as lipid metabolism, glycosylation and vesicle trafficking, investigators are often faced with the challenge of determining the degree to which these pathways are functionally conserved in zebrafish. While they share a high degree of genetic homology with mouse and human, the manner in which cellular pathways are regulated in zebrafish during early development, and the differences in the organ physiology, warrant consideration before functional studies can be effectively interpreted and compared with other vertebrate systems. This point is particularly relevant for glycosylation since an understanding of the glycan diversity and the mechanisms that control glycan biosynthesis during zebrafish embryogenesis (as in many organisms) is still developing.Nonetheless, a growing number of studies in zebrafish have begun to cast light on the functional roles of specific classes of glycans during organ and tissue development. While many of the initial efforts involved characterizing identified mutants in a number of glycosylation pathways, the use of reverse genetic approaches to directly model glycosylation-related disorders is now increasingly popular. In this review, the glycomics of zebrafish and the developmental expression of their glycans will be briefly summarized along with recent chemical biology approaches to visualize certain classes of glycans within developing embryos. Work regarding the role of protein-bound glycans and glycosaminoglycans (GAG) in zebrafish development and organogenesis will also be highlighted. Lastly, future opportunities and challenges in the expanding field of zebrafish glycobiology are discussed.


Asunto(s)
Desarrollo Embrionario/genética , Glicosaminoglicanos/metabolismo , Polisacáridos , Pez Cebra , Animales , Glicómica , Glicosilación , Humanos , Ratones , Organogénesis , Polisacáridos/genética , Polisacáridos/metabolismo , Unión Proteica , Pez Cebra/crecimiento & desarrollo , Pez Cebra/metabolismo
16.
Exp Cell Res ; 317(16): 2342-52, 2011 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-21722633

RESUMEN

Defects in subunits of the conserved oligomeric Golgi (COG) complex represent a growing subset of congenital disorders of glycosylation (CDGs). In addition to altered protein glycosylation and vesicular trafficking, Cog-deficient patient fibroblasts exhibit a striking delay in the Golgi-disrupting effects of brefeldin A (BFA). Despite the diagnostic value of this BFA resistance, the molecular basis of this response is not known. To investigate potential mechanisms of resistance, we analyzed the localization of the large ARF-GEF, GBF1, in several Cog-deficient cell lines. Our results revealed mislocalization of GBF1 to non-Golgi compartments, in particular the ERGIC, within these cells. Biochemical analysis of GBF1 in control and BFA-treated fibroblasts demonstrated that the steady-state level and membrane recruitment is not substantially affected by COG deficiency, supporting a role for the COG complex in the localization but not membrane association of GBF1. We also showed that pretreatment of fibroblasts with bafilomycin resulted in a GBF1-independent BFA resistance that appears additive with the resistance associated with COG deficiency. These data provide new insight into the mechanism of BFA resistance in Cog-deficient cells by suggesting a role for impaired ARF-GEF localization.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/deficiencia , Brefeldino A/farmacología , Resistencia a Medicamentos/fisiología , Factores de Intercambio de Guanina Nucleótido/metabolismo , Transporte de Proteínas/fisiología , Proteínas Adaptadoras del Transporte Vesicular/genética , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Animales , Células CHO , Cricetinae , Cricetulus , Citoplasma/metabolismo , Retículo Endoplásmico/metabolismo , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Aparato de Golgi/efectos de los fármacos , Aparato de Golgi/metabolismo , Proteínas de la Matriz de Golgi , Células HeLa , Proteínas de Choque Térmico/metabolismo , Humanos , Membranas Intracelulares/metabolismo , Macrólidos/farmacología , Lectinas de Unión a Manosa/metabolismo , Proteínas de la Membrana/metabolismo , Transporte de Proteínas/efectos de los fármacos , ATPasas de Translocación de Protón/antagonistas & inhibidores , ATPasas de Translocación de Protón/metabolismo , Piridinas/farmacología , Quinazolinas , Quinolinas/farmacología , ARN Interferente Pequeño/genética , Tirfostinos/farmacología
17.
J Biol Chem ; 286(28): 24855-64, 2011 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-21613225

RESUMEN

In addition to soluble acid hydrolases, many nonlysosomal proteins have been shown to bear mannose 6-phosphate (Man-6-P) residues. Quantification of the extent of mannose phosphorylation and the relevance to physiological function, however, remain poorly defined. In this study, we investigated the mannose phosphorylation status of leukemia inhibitory factor (LIF), a previously identified high affinity ligand for the cation-independent mannose 6-phosphate receptor (CI-MPR), and we analyzed the effects of this modification on its secretion and uptake in cultured cells. When media from LIF-overexpressing cells were fractionated using a CI-MPR affinity column, 35-45% of the total LIF molecules were bound and specifically eluted with free Man-6-P thus confirming LIF as a bona fide Man-6-P-modified protein. Surprisingly, mass spectrometric analysis of LIF glycopeptides enriched on the CI-MPR column revealed that all six N-glycan sites could be Man-6-P-modified. The relative utilization of these sites, however, was not uniform. Analysis of glycan-deleted LIF mutants demonstrated that loss of glycans bearing the majority of Man-6-P residues leads to higher steady-state levels of secreted LIF. Using mouse embryonic stem cells, we showed that the mannose phosphorylation of LIF mediates its internalization thereby reducing extracellular levels and stimulating embryonic stem cell differentiation. Finally, immunofluorescence experiments indicate that LIF is targeted directly to lysosomes following its biosynthesis, providing another mechanism whereby mannose phosphorylation serves to control extracellular levels of LIF. Failure to modify LIF in the context of mucolipidosis II and its subsequent accumulation in the extracellular space may have important implications for disease pathogenesis.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Embrionarias/metabolismo , Factor Inhibidor de Leucemia/metabolismo , Lisosomas/metabolismo , Manosa/metabolismo , Animales , Células CHO , Cricetinae , Cricetulus , Células Madre Embrionarias/citología , Glicosilación , Células HeLa , Humanos , Factor Inhibidor de Leucemia/genética , Factor Inhibidor de Leucemia/aislamiento & purificación , Lisosomas/genética , Manosa/genética , Manosafosfatos/química , Espectrometría de Masas , Ratones , Mucolipidosis/genética , Mucolipidosis/metabolismo , Mutación , Transporte de Proteínas/fisiología
18.
Hum Gene Ther ; 22(3): 293-301, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20858137

RESUMEN

Fabry disease is caused by an X-linked deficiency of the lysosomal enzyme α-galactosidase A (GLA) and has been treated successfully with enzyme replacement therapy (ERT). Gene therapy has been proposed as an alternative to ERT due to the presumed advantages of continuous, endogenous production of the therapeutic enzyme. GLA production in the liver and its therapeutic efficacy in the Fabry mouse have been demonstrated previously with various viral vector systems. In consideration of the potential advantages of using the salivary glands as endogenous GLA biosynthesis sites, we explored the feasibility of this approach in the Fabry mouse. GLA -/0 or -/- mice received an adenoviral vector (2 × 10(10) or 1 × 10(9) viral particles) expressing GLA to the right submandibular gland via oral cannulation of the submandibular duct. Four days later, animals were sacrificed; saliva, plasma, kidney, liver, and brain were collected and assayed using ELISA, Western blot, and a GLA enzymatic activity assay using both traditional fluorescence methods and isotope dilution mass spectrometry by following the U.S. EPA Method 6800. GLA activity was significantly elevated in the serum and liver of both treatment groups, and improvement in the kidney was marginally significant (P < 0.069) in the high-dose group. Notably, we found that liver and salivary gland produce different glycoforms of the GLA transgene. Only small numbers of adenoviral genomes were observed in the livers of treated animals, but in four of 14 in the high-dose groups, liver levels of adenovirus exceeded 20 copies/µg, indicating that the sequestration in the salivary gland was imperfect at high doses. Taken together, these results indicate that the salivary gland-based gene therapy for Fabry disease is promising, and further studies with advanced viral vector gene delivery systems (e.g., adeno-associated virus) for long-term treatment appear to be warranted.


Asunto(s)
Enfermedad de Fabry/enzimología , Enfermedad de Fabry/terapia , Regulación Enzimológica de la Expresión Génica , Terapia Genética , Glándulas Salivales/enzimología , alfa-Galactosidasa/genética , alfa-Galactosidasa/metabolismo , Adenoviridae/genética , Adenoviridae/metabolismo , Animales , Modelos Animales de Enfermedad , Femenino , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Células HEK293 , Humanos , Hígado/metabolismo , Hígado/virología , Masculino , Ratones , Ratones Noqueados , Transporte de Proteínas , Saliva/enzimología , Glándulas Salivales/virología , Vías Secretoras/genética , Factores Sexuales , Transgenes/genética , Transgenes/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA