Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 907
Filtrar
1.
Mol Biol Cell ; 35(8): mr6, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-38888935

RESUMEN

Maintenance of a pool of active lysosomes with acidic pH and degradative hydrolases is crucial for cell health. Abnormalities in lysosomal function are closely linked to diseases, such as lysosomal storage disorders, neurodegeneration, intracellular infections, and cancer among others. Emerging body of research suggests the malfunction of lysosomal hydrolase trafficking pathway to be a common denominator of several disease pathologies. However, available conventional tools to assess lysosomal hydrolase trafficking are insufficient and fail to provide a comprehensive picture about the trafficking flux and location of lysosomal hydrolases. To address some of the shortcomings, we designed a genetically-encoded fluorescent reporter containing a lysosomal hydrolase tandemly tagged with pH sensitive and insensitive fluorescent proteins, which can spatiotemporally trace the trafficking of lysosomal hydrolases. As a proof of principle, we demonstrate that the reporter can detect perturbations in hydrolase trafficking, that are induced by pharmacological manipulations and pathophysiological conditions like intracellular protein aggregates. This reporter can effectively serve as a probe for mapping the mechanistic intricacies of hydrolase trafficking pathway in health and disease and is a utilitarian tool to identify genetic and pharmacological modulators of this pathway, with potential therapeutic implications.


Asunto(s)
Hidrolasas , Lisosomas , Manosafosfatos , Transporte de Proteínas , Humanos , Lisosomas/metabolismo , Manosafosfatos/metabolismo , Hidrolasas/metabolismo , Hidrolasas/genética , Fluorescencia , Genes Reporteros , Proteínas Luminiscentes/metabolismo , Proteínas Luminiscentes/genética , Proteínas Fluorescentes Verdes/metabolismo , Concentración de Iones de Hidrógeno , Células HeLa
2.
Mol Genet Metab ; 142(2): 108487, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38733638

RESUMEN

Phosphomannomutase 2 (PMM2) converts mannose-6-phospahate to mannose-1-phosphate; the substrate for GDP-mannose, a building block of the glycosylation biosynthetic pathway. Pathogenic variants in the PMM2 gene have been shown to be associated with protein hypoglycosylation causing PMM2-congenital disorder of glycosylation (PMM2-CDG). While mannose supplementation improves glycosylation in vitro, but not in vivo, we hypothesized that liposomal delivery of mannose-1-phosphate could increase the stability and delivery of the activated sugar to enter the targeted compartments of cells. Thus, we studied the effect of liposome-encapsulated mannose-1-P (GLM101) on global protein glycosylation and on the cellular proteome in skin fibroblasts from individuals with PMM2-CDG, as well as in individuals with two N-glycosylation defects early in the pathway, namely ALG2-CDG and ALG11-CDG. We leveraged multiplexed proteomics and N-glycoproteomics in fibroblasts derived from different individuals with various pathogenic variants in PMM2, ALG2 and ALG11 genes. Proteomics data revealed a moderate but significant change in the abundance of some of the proteins in all CDG fibroblasts upon GLM101 treatment. On the other hand, N-glycoproteomics revealed the GLM101 treatment enhanced the expression levels of several high-mannose and complex/hybrid glycopeptides from numerous cellular proteins in individuals with defects in PMM2 and ALG2 genes. Both PMM2-CDG and ALG2-CDG exhibited several-fold increase in glycopeptides bearing Man6 and higher glycans and a decrease in Man5 and smaller glycan moieties, suggesting that GLM101 helps in the formation of mature glycoforms. These changes in protein glycosylation were observed in all individuals irrespective of their genetic variants. ALG11-CDG fibroblasts also showed increase in high mannose glycopeptides upon treatment; however, the improvement was not as dramatic as the other two CDG. Overall, our findings suggest that treatment with GLM101 overcomes the genetic block in the glycosylation pathway and can be used as a potential therapy for CDG with enzymatic defects in early steps in protein N-glycosylation.


Asunto(s)
Trastornos Congénitos de Glicosilación , Fibroblastos , Liposomas , Manosafosfatos , Fosfotransferasas (Fosfomutasas) , Humanos , Glicosilación/efectos de los fármacos , Trastornos Congénitos de Glicosilación/genética , Trastornos Congénitos de Glicosilación/tratamiento farmacológico , Trastornos Congénitos de Glicosilación/metabolismo , Trastornos Congénitos de Glicosilación/patología , Fibroblastos/metabolismo , Fibroblastos/efectos de los fármacos , Manosafosfatos/metabolismo , Fosfotransferasas (Fosfomutasas)/genética , Fosfotransferasas (Fosfomutasas)/metabolismo , Fosfotransferasas (Fosfomutasas)/deficiencia , Proteómica , Manosa/metabolismo
4.
J Pharmacol Exp Ther ; 389(3): 313-314, 2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38772716

RESUMEN

We thank Dr. Weimer and her colleagues for their comments related to our recent work (Anding et al., 2023) and are grateful for the opportunity to further discuss the importance of efficient lysosomal targeting of enzyme-replacement therapies (ERT) for the treatment of Pompe disease. Patients with Pompe disease have mutations in the gene that encodes for acid α glucosidase (GAA), a lysosomal enzyme necessary for the breakdown of glycogen. The first-generation ERT, alglucosidase alfa, provides a lifesaving therapy for the severe form of the disease (infantile onset Pompe disease) and improves or stabilizes respiratory and motor function in patients with less severe disease (late onset Pompe disease). Despite these gains, significant unmet need remains, particularly in patients who display respiratory and motor decline following years of treatment. Poor tissue uptake and lysosomal targeting via inefficient binding of the cation-independent mannose-6-phosphate (M6P) receptor (CIMPR) in skeletal muscle contributed to this suboptimal treatment response, prompting the development of new ERTs with increased levels of M6P.


Asunto(s)
1-Desoxinojirimicina , Terapia de Reemplazo Enzimático , Enfermedad del Almacenamiento de Glucógeno Tipo II , Manosafosfatos , alfa-Glucosidasas , Enfermedad del Almacenamiento de Glucógeno Tipo II/tratamiento farmacológico , Animales , Terapia de Reemplazo Enzimático/métodos , Manosafosfatos/metabolismo , Ratones , alfa-Glucosidasas/uso terapéutico , alfa-Glucosidasas/metabolismo , alfa-Glucosidasas/administración & dosificación , 1-Desoxinojirimicina/análogos & derivados , 1-Desoxinojirimicina/administración & dosificación , 1-Desoxinojirimicina/uso terapéutico , Humanos , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo
5.
Enzyme Microb Technol ; 177: 110427, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38518553

RESUMEN

d-mannose has been widely used in food, medicine, cosmetic, and food-additive industries. To date, chemical synthesis or enzymatic conversion approaches based on iso/epimerization reactions for d-mannose production suffered from low conversion rate due to the reaction equilibrium, necessitating intricate separation processes for obtaining pure products on an industrial scale. To circumvent this challenge, this study showcased a new approach for d-mannose synthesis from glucose through constructing a phosphorylation-dephosphorylation pathway in an engineered strain. Specifically, the gene encoding phosphofructokinase (PfkA) in glycolytic pathway was deleted in Escherichia coli to accumulate fructose-6-phosphate (F6P). Additionally, one endogenous phosphatase, YniC, with high specificity to mannose-6-phosphate, was identified. In ΔpfkA strain, a recombinant synthetic pathway based on mannose-6-phosphate isomerase and YniC was developed to direct F6P to mannose. The resulting strain successfully produced 25.2 g/L mannose from glucose with a high conversion rate of 63% after transformation for 48 h. This performance surpassed the 15% conversion rate observed with 2-epimerases. In conclusion, this study presents an efficient method for achieving high-yield mannose synthesis from cost-effective glucose.


Asunto(s)
Escherichia coli , Glucosa , Manosa , Manosa/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Fosforilación , Glucosa/metabolismo , Proteínas de Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Manosafosfatos/metabolismo , Ingeniería Metabólica , Fructosafosfatos/metabolismo , Manosa-6-Fosfato Isomerasa/metabolismo , Manosa-6-Fosfato Isomerasa/genética , Monoéster Fosfórico Hidrolasas/metabolismo , Monoéster Fosfórico Hidrolasas/genética , Glucólisis
6.
Front Cell Infect Microbiol ; 14: 1349221, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38357444

RESUMEN

Viruses, despite their simple structural composition, engage in intricate and complex interactions with their hosts due to their parasitic nature. A notable demonstration of viral behavior lies in their exploitation of lysosomes, specialized organelles responsible for the breakdown of biomolecules and clearance of foreign substances, to bolster their own replication. The man-nose-6-phosphate (M6P) pathway, crucial for facilitating the proper transport of hydrolases into lysosomes and promoting lysosome maturation, is frequently exploited for viral manipulation in support of replication. Recently, the discovery of lysosomal enzyme trafficking factor (LYSET) as a pivotal regulator within the lysosomal M6P pathway has introduced a fresh perspective on the intricate interplay between viral entry and host factors. This groundbreaking revelation illuminates unexplored dimensions of these interactions. In this review, we endeavor to provide a thorough overview of the M6P pathway and its intricate interplay with viral factors during infection. By consolidating the current understanding in this field, our objective is to establish a valuable reference for the development of antiviral drugs that selectively target the M6P pathway.


Asunto(s)
Hidrolasas , Virosis , Humanos , Hidrolasas/metabolismo , Manosafosfatos/análisis , Manosafosfatos/química , Manosafosfatos/metabolismo , Virosis/metabolismo , Lisosomas/metabolismo
7.
Biomater Sci ; 11(5): 1810-1827, 2023 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-36655818

RESUMEN

Stimuli-responsive cross-linked nanocarriers that can induce lysosomal cell death (LCD) via lysosomal membrane permeabilization (LMP) represent a new class of delivery platforms and have attracted the attention of researchers in the biomedical field. The advantages of such cross-linked nanocarriers are as follows (i) they remain intact during blood circulation; and (ii) they reach the target site via specific receptor-mediated endocytosis leading to the enhancement of therapeutic efficacy and reduction of side effects. Herein, we have synthesized a mannose-6-phosphate (M6P) based amphiphilic ABC type tri-block copolymer having two chains of FDA-approved poly(ε-caprolactone) (PCL) as the hydrophobic block, and poly(S-(o-nitrobenzyl)-L-cysteine) (NBC) acts as the photoresponsive crosslinker block. Two different tri-block copolymers, [(PCL35)2-b-NBC20-b-M6PGP20] and [(PCL35)2-b-NBC15-b-M6PGP20], were synthesized which upon successful self-assembly initially formed spherical uncross-linked "micellar-type" aggregates (UCL-M) and vesicles (UCL-V), respectively. The uncross-linked nanocarriers upon UV treatment for thirty minutes were covalently crosslinked in the middle PNBC block giving rise to the di-sulfide bonds and forming interface cross-linked "micellar-type" aggregates (ICL-M) and vesicles (ICL-V). DLS, TEM, and AFM techniques were used to successfully characterize the morphology of these nanocarriers. The dual stimuli (redox and enzyme) responsiveness of the cross-linked nanocarriers and their trafficking to the lysosome in mammalian cells via receptor-mediated endocytosis was probed using confocal microscopy images. Furthermore, the addition of a chloroquine (CQ, a known lysosomotropic agent) encapsulated cross-linked nanocarrier (CQ@ICL-V) to non-cancerous (HEK-293T) cells and liver (HepG2), and breast cancer cells (MDA-MB-231) was found to initiate lysosomal membrane permeabilization (LMP) followed by lysosomal destabilization which eventually led to lysosomal cell death (LCD). Due to the targeted delivery of CQ to the lysosomes of cancerous cells, almost a 90% smaller amount of CQ was able to achieve similar cell death to CQ alone.


Asunto(s)
Manosafosfatos , Polímeros , Animales , Polímeros/química , Manosafosfatos/metabolismo , Micelas , Lisosomas/metabolismo , Mamíferos
8.
Autophagy ; 19(5): 1596-1598, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36633445

RESUMEN

Vertebrate cells rely on mannose-6-phosphate (M6P) modifications to deliver most lumenal hydrolases to the lysosome. As a critical trafficking signal for lysosomal enzymes, the M6P biosynthetic pathway has been thoroughly investigated. However, its regulatory mechanism is largely unknown. Here, we summarize three recent studies that independently discovered LYSET/TMEM251/GCAF as a key regulator of the M6P pathway. LYSET/TMEM251 directly interacts with GNPT, the enzyme that catalyzes the transfer of M6P, and is critical for its activity and stability. Deleting LYSET/TMEM251 impairs the GNPT function and M6P modifications. Consequently, lysosomal enzymes are mistargeted for secretion. Defective lysosomes fail to degrade cargoes such as endocytic vesicles and autophagosomes, leading to a newly identified lysosomal storage disease in humans. These discoveries open up a new direction in the regulation of the M6P biosynthetic pathway.Abbreviations: ER: endoplasmic reticulum; GNPT: GlcNAc-1-phosphotransferase; KO: knockout; LMP: lysosome membrane protein; LYSET: lysosomal enzyme trafficking factor; MAP1LC3/LC3: microtubule-associated protein 1 light chain 3; M6P: mannose-6-phosphate; MBTPS1/S1P: membrane-bound transcription factor peptidase, site 1; MPR: mannose-6-phosphate receptor; SQSTM1: sequestosome 1; TEM: transmission electron microscopy; TGN: trans-Golgi network.


Asunto(s)
Autofagia , Enfermedades por Almacenamiento Lisosomal , Humanos , Lisosomas/metabolismo , Enfermedades por Almacenamiento Lisosomal/metabolismo , Manosafosfatos/metabolismo , Hidrolasas/metabolismo
9.
Commun Biol ; 6(1): 48, 2023 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-36639722

RESUMEN

N-glycosylation is implicated in cancers and aberrant N-glycosylation is recognized as a hallmark of cancer. Here, we mapped and compared the site-specific N-glycoproteomes of colon cancer HCT116 cells and isogenic non-tumorigenic DNMT1/3b double knockout (DKO1) cells using Fbs1-GYR N-glycopeptide enrichment technology and trapped ion mobility spectrometry. Many significant changes in site-specific N-glycosylation were revealed, providing a molecular basis for further elucidation of the role of N-glycosylation in protein function. HCT116 cells display hypersialylation especially in cell surface membrane proteins. Both HCT116 and DKO1 show an abundance of paucimannose and 80% of paucimannose-rich proteins are annotated to reside in exosomes. The most striking N-glycosylation alteration was the degree of mannose-6-phosphate (M6P) modification. N-glycoproteomic analyses revealed that HCT116 displays hyper-M6P modification, which was orthogonally validated by M6P immunodetection. Significant observed differences in N-glycosylation patterns of the major M6P receptor, CI-MPR in HCT116 and DKO1 may contribute to the hyper-M6P phenotype of HCT116 cells. This comparative site-specific N-glycoproteome analysis provides a pool of potential N-glycosylation-related cancer biomarkers, but also gives insights into the M6P pathway in cancer.


Asunto(s)
Manosafosfatos , Neoplasias , Humanos , Glicosilación , Manosafosfatos/química , Manosafosfatos/metabolismo , Neoplasias/genética
10.
Nat Commun ; 13(1): 5351, 2022 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-36096887

RESUMEN

The mannose-6-phosphate (M6P) biosynthetic pathway for lysosome biogenesis has been studied for decades and is considered a well-understood topic. However, whether this pathway is regulated remains an open question. In a genome-wide CRISPR/Cas9 knockout screen, we discover TMEM251 as the first regulator of the M6P modification. Deleting TMEM251 causes mistargeting of most lysosomal enzymes due to their loss of M6P modification and accumulation of numerous undigested materials. We further demonstrate that TMEM251 localizes to the Golgi and is required for the cleavage and activity of GNPT, the enzyme that catalyzes M6P modification. In zebrafish, TMEM251 deletion leads to severe developmental defects including heart edema and skeletal dysplasia, which phenocopies Mucolipidosis Type II. Our discovery provides a mechanism for the newly discovered human disease caused by TMEM251 mutations. We name TMEM251 as GNPTAB cleavage and activity factor (GCAF) and its related disease as Mucolipidosis Type V.


Asunto(s)
Proteínas de la Membrana , Mucolipidosis , Pez Cebra , Animales , Humanos , Lisosomas/metabolismo , Manosafosfatos/metabolismo , Proteínas de la Membrana/metabolismo , Mucolipidosis/genética , Mucolipidosis/metabolismo , Transferasas (Grupos de Otros Fosfatos Sustitutos)/genética , Transferasas (Grupos de Otros Fosfatos Sustitutos)/metabolismo , Pez Cebra/metabolismo
11.
Proc Natl Acad Sci U S A ; 119(33): e2203518119, 2022 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-35939698

RESUMEN

The mannose-6-phosphate (M6P) pathway is responsible for the transport of hydrolytic enzymes to lysosomes. N-acetylglucosamine-1-phosphotransferase (GNPT) catalyzes the first step of tagging these hydrolases with M6P, which when recognized by receptors in the Golgi diverts them to lysosomes. Genetic defects in the GNPT subunits, GNPTAB and GNPTG, cause the lysosomal storage diseases mucolipidosis types II and III. To better understand its function, we determined partial three-dimensional structures of the GNPT complex. The catalytic domain contains a deep cavity for binding of uridine diphosphate-N-acetylglucosamine, and the surrounding residues point to a one-step transfer mechanism. An isolated structure of the gamma subunit of GNPT reveals that it can bind to mannose-containing glycans in different configurations, suggesting that it may play a role in directing glycans into the active site. These findings may facilitate the development of therapies for lysosomal storage diseases.


Asunto(s)
Enfermedades por Almacenamiento Lisosomal , Manosafosfatos , Mucolipidosis , Transferasas (Grupos de Otros Fosfatos Sustitutos) , Dominio Catalítico , Humanos , Enfermedades por Almacenamiento Lisosomal/metabolismo , Lisosomas/enzimología , Manosafosfatos/metabolismo , Mucolipidosis/enzimología , Transferasas (Grupos de Otros Fosfatos Sustitutos)/química , Transferasas (Grupos de Otros Fosfatos Sustitutos)/genética
12.
Plant Cell Physiol ; 63(5): 658-670, 2022 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-35243499

RESUMEN

Sugar alcohols are major photosynthetic products in plant species from the Apiaceae and Plantaginaceae families. Mannose-6-phosphate reductase (Man6PRase) and aldose-6-phosphate reductase (Ald6PRase) are key enzymes for synthesizing mannitol and glucitol in celery (Apium graveolens) and peach (Prunus persica), respectively. In this work, we report the first crystal structures of dimeric plant aldo/keto reductases (AKRs), celery Man6PRase (solved in the presence of mannonic acid and NADP+) and peach Ald6PRase (obtained in the apo form). Both structures displayed the typical TIM barrel folding commonly observed in proteins from the AKR superfamily. Analysis of the Man6PRase holo form showed that residues putatively involved in the catalytic mechanism are located close to the nicotinamide ring of NADP+, where the hydride transfer to the sugar phosphate should take place. Additionally, we found that Lys48 is important for the binding of the sugar phosphate. Interestingly, the Man6PRase K48A mutant had a lower catalytic efficiency with mannose-6-phosphate but a higher catalytic efficiency with mannose than the wild type. Overall, our work sheds light on the structure-function relationships of important enzymes to synthesize sugar alcohols in plants.


Asunto(s)
Fosfatos , Alcoholes del Azúcar , Oxidorreductasas de Alcohol/metabolismo , Aldehído Reductasa/metabolismo , Secuencia de Aminoácidos , Humanos , Manosafosfatos , NADP/metabolismo , Plantas/metabolismo , Azúcares
13.
Carbohydr Res ; 511: 108489, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34922155

RESUMEN

Currently, the reaction toolbox for the functionalization of glycans assembled on solid-phase is quite limited. Automated (1 h) and manual (overnight) phosphorylation protocols that enable the solid-phase synthesis of oligosaccharides containing up to two mannose-6-phosphates are presented. Automated glycan assembly expedited access to substrates and facilitated the screening of experimental conditions.


Asunto(s)
Manosafosfatos , Oligosacáridos , Manosa , Polisacáridos , Técnicas de Síntesis en Fase Sólida
14.
Biochem Biophys Res Commun ; 579: 54-61, 2021 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-34587555

RESUMEN

1,2-ß-Mannobiose phosphorylases (1,2-ß-MBPs) from glycoside hydrolase 130 (GH130) family are important bio-catalysts in glycochemistry applications owing to their ability in synthesizing oligomannans. Here, we report the crystal structure of a thermostable 1,2-ß-MBP from Thermoanaerobacter sp. X-514 termed Teth514_1789 to reveal the molecular basis of its higher thermostability and mechanism of action. We also solved the enzyme complexes of mannose, mannose-1-phosphate (M1P) and 1,4-ß-mannobiose to manifest the enzyme-substrate interaction networks of three main subsites. Notably, a Zn ion that should be derived from crystallization buffer was found in the active site and coordinates the phosphate moiety of M1P. Nonetheless, this Zn-coordination should reflect an inhibitory status as supplementing Zn severely impairs the enzyme activity. These results indicate that the effects of metal ions should be taken into consideration when applying Teth514_1789 and other related enzymes. Based on the structure, a reliable model of Teth514_1788 that shares 61.7% sequence identity to Teth514_1789 but displays a different substrate preference was built. Analyzing the structural features of these two closely related enzymes, we hypothesized that the length of a loop fragment that covers the entrance of the catalytic center might regulate the substrate selectivity. In conclusion, these information provide in-depth understanding of GH130 1,2-ß-MBPs and should serve as an important guidance for enzyme engineering for further applications.


Asunto(s)
Thermoanaerobacter/enzimología , beta-Manosidasa/química , Sitios de Unión , Catálisis , Dominio Catalítico , Glicósido Hidrolasas/química , Iones , Ligandos , Mananos/química , Manosa/química , Manosafosfatos/química , Fosforilasas/química , Plásmidos/metabolismo , Conformación Proteica , Reproducibilidad de los Resultados , Electricidad Estática , Temperatura , Zinc/química
15.
Anal Bioanal Chem ; 413(29): 7295-7303, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34155551

RESUMEN

The coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) presents a serious threat to human health all over the world. The development of effective vaccines has been focusing on the spike (S) glycoprotein, which mediates viral invasion to human cells through its interaction with the angiotensin-converting enzyme 2 (ACE2) receptor. In this work, we perform analytical characterization of N- and O-linked glycosylation of the SARS-CoV-2 S glycoprotein. We explore the novel use of dual-functionalized titanium (IV)-immobilized metal affinity chromatography (Ti-IMAC) material for simultaneous enrichment and separation of neutral and sialyl glycopeptides of a recombinant SARS-CoV-2 S glycoprotein from HEK293 cells. This strategy helps eliminate signal suppression from neutral glycopeptides for the detection of sialyl glycopeptides and improves the glycoform coverage of the S protein. We profiled 19 of its 22 potential N-glycosylated sites with 398 unique glycoforms using the dual-functional Ti-IMAC approach, which exhibited improvement of coverage by 1.6-fold compared to the conventional hydrophilic interaction chromatography (HILIC) glycopeptide enrichment method. We also identified O-linked glycosylation site that was not found using the conventional HILIC approach. In addition, we reported on the identification of mannose-6-phosphate (M6P) glycosylation, which substantially expands the current knowledge of the spike protein's glycosylation landscape and enables future investigation into the influence of M6P glycosylation of the spike protein on its cell entry.


Asunto(s)
Glicopéptidos/aislamiento & purificación , Ácido N-Acetilneuramínico/química , SARS-CoV-2/química , Glicoproteína de la Espiga del Coronavirus/química , Secuencia de Aminoácidos , Cromatografía Liquida/métodos , Glicopéptidos/química , Células HEK293 , Humanos , Manosafosfatos/química , Electricidad Estática , Espectrometría de Masas en Tándem/métodos
16.
J Clin Invest ; 131(15)2021 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-34128834

RESUMEN

Disordered lysosomal/autophagy pathways initiate and drive pancreatitis, but the underlying mechanisms and links to disease pathology are poorly understood. Here, we show that the mannose-6-phosphate (M6P) pathway of hydrolase delivery to lysosomes critically regulates pancreatic acinar cell cholesterol metabolism. Ablation of the Gnptab gene encoding a key enzyme in the M6P pathway disrupted acinar cell cholesterol turnover, causing accumulation of nonesterified cholesterol in lysosomes/autolysosomes, its depletion in the plasma membrane, and upregulation of cholesterol synthesis and uptake. We found similar dysregulation of acinar cell cholesterol, and a decrease in GNPTAB levels, in both WT experimental pancreatitis and human disease. The mechanisms mediating pancreatic cholesterol dyshomeostasis in Gnptab-/- and experimental models involve a disordered endolysosomal system, resulting in impaired cholesterol transport through lysosomes and blockage of autophagic flux. By contrast, in Gnptab-/- liver the endolysosomal system and cholesterol homeostasis were largely unaffected. Gnptab-/- mice developed spontaneous pancreatitis. Normalization of cholesterol metabolism by pharmacologic means alleviated responses of experimental pancreatitis, particularly trypsinogen activation, the disease hallmark. The results reveal the essential role of the M6P pathway in maintaining exocrine pancreas homeostasis and function, and implicate cholesterol disordering in the pathogenesis of pancreatitis.


Asunto(s)
Células Acinares/metabolismo , Colesterol/metabolismo , Manosafosfatos/metabolismo , Páncreas Exocrino/metabolismo , Pancreatitis/metabolismo , Células Acinares/patología , Animales , Colesterol/genética , Modelos Animales de Enfermedad , Humanos , Manosafosfatos/genética , Ratones , Ratones Noqueados , Páncreas Exocrino/patología , Pancreatitis/patología , Transferasas (Grupos de Otros Fosfatos Sustitutos)/deficiencia , Transferasas (Grupos de Otros Fosfatos Sustitutos)/metabolismo
17.
FEBS Open Bio ; 11(6): 1695-1703, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33932147

RESUMEN

Disruption of the mannose 6-phosphate (M-6-P) pathway in HeLa cells by inactivation of the GNPTAB gene, which encodes the α/ß subunits of GlcNAc-1-phosphotransferase, results in missorting of newly synthesized lysosomal acid hydrolases to the cell culture media instead of transport to the endolysosomal system. We previously demonstrated that the majority of the lysosomal aspartyl protease, cathepsin D, is secreted in these GNPTAB-/- HeLa cells. However, the intracellular content of cathepsin D in these cells was still greater than that of WT HeLa cells which retained most of the protease, indicating a marked elevation of cathepsin D expression in response to abrogation of the M-6-P pathway. Here, we demonstrate that HeLa cells lacking GlcNAc-1-phosphotransferase show a fivefold increase in cathepsin D mRNA expression over control cells, accounting for the increase in cathepsin D at the protein level. Further, we show that this increase at the mRNA level occurs independent of the transcription factors TFEB and TFE3. The intracellular cathepsin D can still be trafficked to lysosomes in the absence of the M-6-P pathway, but fails to undergo proteolytic processing into the fully mature heavy and light chains. Uptake experiments performed by feeding GNPTAB-/- HeLa cells with various phosphorylated cathepsins reveal that only cathepsin B is capable of partially restoring cleavage, providing evidence for a role for cathepsin B in the proteolytic processing of cathepsin D.


Asunto(s)
Catepsina D/genética , ARN Mensajero/genética , Catepsina D/metabolismo , Células HeLa , Humanos , Manosafosfatos/metabolismo , ARN Mensajero/metabolismo , Transferasas (Grupos de Otros Fosfatos Sustitutos)/deficiencia , Transferasas (Grupos de Otros Fosfatos Sustitutos)/metabolismo
18.
Sci Rep ; 11(1): 8213, 2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33859256

RESUMEN

Plasmin is the key enzyme in fibrinolysis. Upon interaction with plasminogen activators, the zymogen plasminogen is converted to active plasmin. Some studies indicate plasminogen activation is regulated by cation-independent mannose 6-phosphate receptor (CI-MPR), a protein that facilitates lysosomal enzyme trafficking and insulin-like growth factor 2 downregulation. Plasminogen regulation may be accomplished by CI-MPR binding to plasminogen or urokinase plasminogen activator receptor. We asked whether other members of the plasminogen activation system, such as tissue plasminogen activator (tPA), also interact with CI-MPR. Because tPA is a glycoprotein with three N-linked glycosylation sites, we hypothesized that tPA contains mannose 6-phosphate (M6P) and binds CI-MPR in a M6P-dependent manner. Using surface plasmon resonance, we found that two sources of tPA bound the extracellular region of human and bovine CI-MPR with low-mid nanomolar affinities. Binding was partially inhibited with phosphatase treatment or M6P. Subsequent studies revealed that the five N-terminal domains of CI-MPR were sufficient for tPA binding, and this interaction was also partially mediated by M6P. The three glycosylation sites of tPA were analyzed by mass spectrometry, and glycoforms containing M6P and M6P-N-acetylglucosamine were identified at position N448 of tPA. In summary, we found that tPA contains M6P and is a CI-MPR ligand.


Asunto(s)
Manosafosfatos/metabolismo , Receptor IGF Tipo 2/metabolismo , Activador de Tejido Plasminógeno/metabolismo , Acetilglucosamina/metabolismo , Animales , Células CHO , Células Cultivadas , Cricetulus , Glicoproteínas/química , Glicoproteínas/metabolismo , Humanos , Factor II del Crecimiento Similar a la Insulina/química , Factor II del Crecimiento Similar a la Insulina/metabolismo , Ligandos , Fosforilación , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Receptor IGF Tipo 2/química , Células Sf9 , Spodoptera , Activador de Tejido Plasminógeno/química , Activador de Tejido Plasminógeno/fisiología
19.
J Cosmet Dermatol ; 20(6): 1598-1610, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33580613

RESUMEN

BACKGROUND: The dermis is composed of a tangle of macromolecules that provides the skin its biomechanical properties. During chronological aging, fibroblasts lose their ability to synthesize collagen and an accumulation of matrix metalloproteinases leads to an increase in collagen degradation. As a result, there is a decline in the biomechanical properties of the skin. Skin aging is accelerated by external factors such as UV radiation and pollution, which induce accumulation of oxidants, and so of oxidized proteins in the skin. AIMS: Atomic force microscopy (AFM) has emerged as an alternative method for studying the biomechanical properties of skin cells and tissues. METHODS/RESULTS: Thus, we identified mannose-6-phosphate complex as a new powerful molecule capable of reversing the visible signs of aging by reorganizing the collagen network of the dermis and by improving the skin biomechanical properties. This effect was correlated with clinical studies that showed a marked antiaging effect through a reduction in the number of crow's feet and in the depth and size of neck wrinkles. CONCLUSION: Mannose-6-phosphate complex appeared to be able to protect proteins in the dermis scaffold against oxidation and degradation, allowing an improvement in the skin biomechanical properties.


Asunto(s)
Envejecimiento de la Piel , Fibroblastos , Manosafosfatos , Piel
20.
J Microbiol Biotechnol ; 31(1): 163-170, 2021 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-33144549

RESUMEN

Enzyme replacement therapy for lysosomal storage diseases usually requires recombinant enzymes containing mannose-6-phosphate (M6P) glycans for cellular uptake and lysosomal targeting. For the first time, a strategy is established here for the in vitro mannosyl-phosphorylation of high-mannose type N-glycans that utilizes a recombinant Mnn14 protein derived from Saccharomyces cerevisiae. Among a series of N-terminal- or C-terminal-deleted recombinant Mnn14 proteins expressed in Pichia pastoris, rMnn1477-935 with deletion of N-terminal 76 amino acids spanning the transmembrane domain (46 amino acids) and part of the stem region (30 amino acids), showed the highest level of mannosyl-phosphorylation activity. The optimum reaction conditions for rMnn1477-935 were determined through enzyme assays with a high-mannose type N-glycan (Man8GlcNAc2) as a substrate. In addition, rMnn1477-935 was shown to mannosyl-phosphorylate high-mannose type Nglycans (Man7-9GlcNAc2) on recombinant human lysosomal alpha-glucosidase (rhGAA) with remarkably high efficiency. Moreover, the majority of the resulting mannosyl-phosphorylated glycans were bis-form which can be converted to bis-phosphorylated M6P glycans having a superior lysosomal targeting capability. An in vitro N-glycan mannosyl-phosphorylation reaction using rMnn1477-935 will provide a flexible and straightforward method to increase the M6P glycan content for the generation of "Biobetter" therapeutic enzymes.


Asunto(s)
Manosafosfatos/metabolismo , Polisacáridos/metabolismo , Proteínas Recombinantes/genética , Saccharomycetales/genética , Saccharomycetales/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Fosforilación , Pichia/metabolismo , Proteínas Recombinantes/química , Saccharomyces cerevisiae , Proteínas de Saccharomyces cerevisiae/genética , Temperatura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA