Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 570
Filter
1.
J Pharmacol Exp Ther ; 389(3): 313-314, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38772716

ABSTRACT

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.


Subject(s)
1-Deoxynojirimycin , Enzyme Replacement Therapy , Glycogen Storage Disease Type II , Mannosephosphates , alpha-Glucosidases , Glycogen Storage Disease Type II/drug therapy , Animals , Enzyme Replacement Therapy/methods , Mannosephosphates/metabolism , Mice , alpha-Glucosidases/therapeutic use , alpha-Glucosidases/metabolism , alpha-Glucosidases/administration & dosage , 1-Deoxynojirimycin/analogs & derivatives , 1-Deoxynojirimycin/administration & dosage , 1-Deoxynojirimycin/therapeutic use , Humans , Lysosomes/drug effects , Lysosomes/metabolism
2.
Enzyme Microb Technol ; 177: 110427, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38518553

ABSTRACT

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.


Subject(s)
Escherichia coli , Glucose , Mannose , Mannose/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Phosphorylation , Glucose/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/genetics , Mannosephosphates/metabolism , Metabolic Engineering , Fructosephosphates/metabolism , Mannose-6-Phosphate Isomerase/metabolism , Mannose-6-Phosphate Isomerase/genetics , Phosphoric Monoester Hydrolases/metabolism , Phosphoric Monoester Hydrolases/genetics , Glycolysis
3.
Front Cell Infect Microbiol ; 14: 1349221, 2024.
Article in English | MEDLINE | ID: mdl-38357444

ABSTRACT

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.


Subject(s)
Hydrolases , Virus Diseases , Humans , Hydrolases/metabolism , Mannosephosphates/analysis , Mannosephosphates/chemistry , Mannosephosphates/metabolism , Virus Diseases/metabolism , Lysosomes/metabolism
4.
Biomater Sci ; 11(5): 1810-1827, 2023 Feb 28.
Article in English | MEDLINE | ID: mdl-36655818

ABSTRACT

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.


Subject(s)
Mannosephosphates , Polymers , Animals , Polymers/chemistry , Mannosephosphates/metabolism , Micelles , Lysosomes/metabolism , Mammals
5.
Autophagy ; 19(5): 1596-1598, 2023 05.
Article in English | MEDLINE | ID: mdl-36633445

ABSTRACT

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.


Subject(s)
Autophagy , Lysosomal Storage Diseases , Humans , Lysosomes/metabolism , Lysosomal Storage Diseases/metabolism , Mannosephosphates/metabolism , Hydrolases/metabolism
6.
Commun Biol ; 6(1): 48, 2023 01 13.
Article in English | MEDLINE | ID: mdl-36639722

ABSTRACT

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.


Subject(s)
Mannosephosphates , Neoplasms , Humans , Glycosylation , Mannosephosphates/chemistry , Mannosephosphates/metabolism , Neoplasms/genetics
7.
Nat Commun ; 13(1): 5351, 2022 09 12.
Article in English | MEDLINE | ID: mdl-36096887

ABSTRACT

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.


Subject(s)
Membrane Proteins , Mucolipidoses , Zebrafish , Animals , Humans , Lysosomes/metabolism , Mannosephosphates/metabolism , Membrane Proteins/metabolism , Mucolipidoses/genetics , Mucolipidoses/metabolism , Transferases (Other Substituted Phosphate Groups)/genetics , Transferases (Other Substituted Phosphate Groups)/metabolism , Zebrafish/metabolism
8.
Proc Natl Acad Sci U S A ; 119(33): e2203518119, 2022 08 16.
Article in English | MEDLINE | ID: mdl-35939698

ABSTRACT

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.


Subject(s)
Lysosomal Storage Diseases , Mannosephosphates , Mucolipidoses , Transferases (Other Substituted Phosphate Groups) , Catalytic Domain , Humans , Lysosomal Storage Diseases/metabolism , Lysosomes/enzymology , Mannosephosphates/metabolism , Mucolipidoses/enzymology , Transferases (Other Substituted Phosphate Groups)/chemistry , Transferases (Other Substituted Phosphate Groups)/genetics
9.
J Clin Invest ; 131(15)2021 08 02.
Article in English | MEDLINE | ID: mdl-34128834

ABSTRACT

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.


Subject(s)
Acinar Cells/metabolism , Cholesterol/metabolism , Mannosephosphates/metabolism , Pancreas, Exocrine/metabolism , Pancreatitis/metabolism , Acinar Cells/pathology , Animals , Cholesterol/genetics , Disease Models, Animal , Humans , Mannosephosphates/genetics , Mice , Mice, Knockout , Pancreas, Exocrine/pathology , Pancreatitis/pathology , Transferases (Other Substituted Phosphate Groups)/deficiency , Transferases (Other Substituted Phosphate Groups)/metabolism
10.
FEBS Open Bio ; 11(6): 1695-1703, 2021 06.
Article in English | MEDLINE | ID: mdl-33932147

ABSTRACT

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.


Subject(s)
Cathepsin D/genetics , RNA, Messenger/genetics , Cathepsin D/metabolism , HeLa Cells , Humans , Mannosephosphates/metabolism , RNA, Messenger/metabolism , Transferases (Other Substituted Phosphate Groups)/deficiency , Transferases (Other Substituted Phosphate Groups)/metabolism
11.
Sci Rep ; 11(1): 8213, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33859256

ABSTRACT

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.


Subject(s)
Mannosephosphates/metabolism , Receptor, IGF Type 2/metabolism , Tissue Plasminogen Activator/metabolism , Acetylglucosamine/metabolism , Animals , CHO Cells , Cells, Cultured , Cricetulus , Glycoproteins/chemistry , Glycoproteins/metabolism , Humans , Insulin-Like Growth Factor II/chemistry , Insulin-Like Growth Factor II/metabolism , Ligands , Phosphorylation , Protein Binding , Protein Interaction Domains and Motifs , Receptor, IGF Type 2/chemistry , Sf9 Cells , Spodoptera , Tissue Plasminogen Activator/chemistry , Tissue Plasminogen Activator/physiology
12.
Chembiochem ; 22(2): 434-440, 2021 01 15.
Article in English | MEDLINE | ID: mdl-32864819

ABSTRACT

Mannose-6-phosphate (M6P) is recognized by the mannose-6-phosphate receptor and plays an important role in the transport of cargo to the endosomes, making it an attractive tool to improve endosomal trafficking of vaccines. We describe herein the assembly of peptide antigen conjugates carrying clusters of mannose-6-C-phosphonates (M6Po). The M6Po's are stable M6P mimics that are resistant to cleavage of the phosphate group by endogenous phosphatases. Two different strategies for the incorporation of the M6Po clusters in the conjugate have been developed: the first relies on a "post-assembly" click approach employing an M6Po bearing an alkyne functionality; the second hinges on an M6Po C-glycoside amino acid building block that can be used in solid-phase peptide synthesis. The generated conjugates were further equipped with a TLR7 ligand to stimulate dendritic cell (DC) maturation. While antigen presentation is hindered by the presence of the M6Po clusters, the incorporation of the M6Po clusters leads to increased activation of DCs, thus demonstrating their potential in improving vaccine adjuvanticity by intraendosomally active TLR ligands.


Subject(s)
Antigens/metabolism , Mannosephosphates/metabolism , Peptides/metabolism , Toll-Like Receptors/metabolism , Antigens/chemistry , Humans , Ligands , Mannosephosphates/chemistry , Molecular Structure , Peptides/chemistry , Toll-Like Receptors/chemistry
13.
J Microbiol Biotechnol ; 31(1): 163-170, 2021 Jan 28.
Article in English | MEDLINE | ID: mdl-33144549

ABSTRACT

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.


Subject(s)
Mannosephosphates/metabolism , Polysaccharides/metabolism , Recombinant Proteins/genetics , Saccharomycetales/genetics , Saccharomycetales/metabolism , Humans , Hydrogen-Ion Concentration , Phosphorylation , Pichia/metabolism , Recombinant Proteins/chemistry , Saccharomyces cerevisiae , Saccharomyces cerevisiae Proteins/genetics , Temperature
14.
Nat Commun ; 11(1): 6343, 2020 12 11.
Article in English | MEDLINE | ID: mdl-33311467

ABSTRACT

D-mannose is a monosaccharide approximately a hundred times less abundant than glucose in human blood. Previous studies demonstrated that supraphysiological levels of D-mannose inhibit tumour growth and stimulate regulatory T cell differentiation. It is not known whether D-mannose metabolism affects the function of non-proliferative cells, such as inflammatory macrophages. Here, we show that D-mannose suppresses LPS-induced macrophage activation by impairing IL-1ß production. In vivo, mannose administration improves survival in a mouse model of LPS-induced endotoxemia as well as decreases progression in a mouse model of DSS-induced colitis. Phosphomannose isomerase controls response of LPS-activated macrophages to D-mannose, which impairs glucose metabolism by raising intracellular mannose-6-phosphate levels. Such alterations result in the suppression of succinate-mediated HIF-1α activation, imposing a consequent reduction of LPS-induced Il1b expression. Disclosing an unrecognized metabolic hijack of macrophage activation, our study points towards safe D-mannose utilization as an effective intervention against inflammatory conditions.


Subject(s)
Interleukin-1beta/metabolism , Macrophage Activation/drug effects , Macrophages/drug effects , Macrophages/metabolism , Mannose/metabolism , Mannose/pharmacology , Animals , Cell Differentiation/drug effects , Cell Line , Colitis/metabolism , Colitis/pathology , Gene Expression Regulation , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Inflammation/metabolism , Interleukin-1beta/genetics , Lipopolysaccharides/adverse effects , Mannosephosphates/metabolism , Metabolic Networks and Pathways/drug effects , Metabolomics , Monocytes/metabolism
15.
Structure ; 28(12): 1300-1312.e5, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32877646

ABSTRACT

The cation-independent mannose 6-phosphate (M6P)/Insulin-like growth factor-2 receptor (CI-MPR/IGF2R) is an ∼300 kDa transmembrane protein responsible for trafficking M6P-tagged lysosomal hydrolases and internalizing IGF2. The extracellular region of the CI-MPR has 15 homologous domains, including M6P-binding domains (D) 3, 5, 9, and 15 and IGF2-binding domain 11. We have focused on solving the first structures of human D7-10 within two multi-domain constructs, D9-10 and D7-11, and provide the first high-resolution description of the high-affinity M6P-binding D9. Moreover, D9 stabilizes a well-defined hub formed by D7-11 whereby two penta-domains intertwine to form a dimeric helical-type coil via an N-glycan bridge on D9. Remarkably the D7-11 structure matches an IGF2-bound state of the receptor, suggesting this may be an intrinsically stable conformation at neutral pH. Interdomain clusters of histidine and proline residues may impart receptor rigidity and play a role in structural transitions at low pH.


Subject(s)
Receptor, IGF Type 2/chemistry , Binding Sites , Humans , Hydrogen-Ion Concentration , Mannosephosphates/chemistry , Mannosephosphates/metabolism , Molecular Docking Simulation , Protein Binding , Protein Multimerization , Receptor, IGF Type 2/metabolism
16.
Int J Mol Sci ; 21(18)2020 Sep 17.
Article in English | MEDLINE | ID: mdl-32957425

ABSTRACT

Mucolipidosis II and III (ML II/III) are caused by a deficiency of uridine-diphosphate N-acetylglucosamine: lysosomal-enzyme-N-acetylglucosamine-1-phosphotransferase (GlcNAc-1-phosphotransferase, EC2.7.8.17), which tags lysosomal enzymes with a mannose 6-phosphate (M6P) marker for transport to the lysosome. The process is performed by a sequential two-step process: first, GlcNAc-1-phosphotransferase catalyzes the transfer of GlcNAc-1-phosphate to the selected mannose residues on lysosomal enzymes in the cis-Golgi network. The second step removes GlcNAc from lysosomal enzymes by N-acetylglucosamine-1-phosphodiester α-N-acetylglucosaminidase (uncovering enzyme) and exposes the mannose 6-phosphate (M6P) residues in the trans-Golgi network, in which the enzymes are targeted to the lysosomes by M6Preceptors. A deficiency of GlcNAc-1-phosphotransferase causes the hypersecretion of lysosomal enzymes out of cells, resulting in a shortage of multiple lysosomal enzymes within lysosomes. Due to a lack of GlcNAc-1-phosphotransferase, the accumulation of cholesterol, phospholipids, glycosaminoglycans (GAGs), and other undegraded substrates occurs in the lysosomes. Clinically, ML II and ML III exhibit quite similar manifestations to mucopolysaccharidoses (MPSs), including specific skeletal deformities known as dysostosis multiplex and gingival hyperplasia. The life expectancy is less than 10 years in the severe type, and there is no definitive treatment for this disease. In this review, we have described the updated diagnosis and therapy on ML II/III.


Subject(s)
Enzyme Replacement Therapy/methods , Genetic Therapy/methods , Hematopoietic Stem Cell Transplantation/methods , Lysosomes/metabolism , Mucolipidoses/diagnosis , Animals , Biological Transport, Active , Disease Models, Animal , Glycosaminoglycans/metabolism , Humans , Mannosephosphates/metabolism , Mucolipidoses/enzymology , Mucolipidoses/physiopathology , Mucolipidoses/therapy
17.
Methods Mol Biol ; 2132: 267-276, 2020.
Article in English | MEDLINE | ID: mdl-32306334

ABSTRACT

Cation-dependent mannose 6-phosphate receptor (CD-MPR) and cation-independent MPR (CI-MPR) belong to the P-type lectin family. Both intracellular and cell surface MPRs can recognize and bind with the terminal mannose 6-phospahte (M6P) residues of N-glycans attached to the mammalian lysosomal enzymes and the related co-factors. Domain9 (Dom9), which is one of the extracytoplasmic region of the CI-MPR, has relatively higher affinity for M6P residues. Here we describe the production of recombinant Dom9-His protein by Pichia pastris, purification, and application as a probe for lectin blotting.


Subject(s)
Pichia/growth & development , Polysaccharides/metabolism , Receptor, IGF Type 2/chemistry , Receptor, IGF Type 2/metabolism , Amino Acid Sequence , Binding Sites , Cloning, Molecular , Humans , Lysosomes/metabolism , Mannosephosphates/metabolism , Pichia/genetics , Pichia/metabolism , Protein Domains , Receptor, IGF Type 2/genetics
18.
Structure ; 28(4): 426-436.e3, 2020 04 07.
Article in English | MEDLINE | ID: mdl-32109365

ABSTRACT

Most lysosomal hydrolytic enzymes reach their destination via the mannose-6-phosphate (M6P) pathway. The enzyme N-acetylglucosamine-1-phosphodiester α-N-acetylglucosaminidase (NAGPA, or "uncovering enzyme") catalyzes the second step in the M6P tag formation, namely the removal of the masking N-acetylglucosamine (GlcNAc) portion. Defects in this protein are associated with non-syndromic stuttering. To gain a better understanding of the function and regulation of this enzyme, we determined its crystal structure. The propeptide binds in a groove on the globular catalytic domain, blocking active site access. High-affinity substrate binding is enabled by a conformational switch in an active site loop. The protein recognizes the GlcNAc and phosphate portions of its substrate, but not the mannose moiety of the glycan. Based on enzymatic and 1H-NMR analysis, a catalytic mechanism is proposed. Crystallographic and solution scattering analyses suggest that the C-terminal domain forms a long flexible stem that extends the enzyme away from the Golgi membrane.


Subject(s)
Catalytic Domain , Phosphoric Diester Hydrolases/chemistry , Acetylglucosamine/chemistry , Acetylglucosamine/metabolism , Animals , Crystallography, X-Ray , Humans , Mannosephosphates/chemistry , Mannosephosphates/metabolism , Phosphoric Diester Hydrolases/metabolism , Protein Binding , Protein Conformation, beta-Strand , Sf9 Cells , Spodoptera
19.
Protein Expr Purif ; 170: 105589, 2020 06.
Article in English | MEDLINE | ID: mdl-32027983

ABSTRACT

The cation-independent mannose-6-phosphate receptor (CI-M6PR, aka insulin-like growth factor II receptor or IGFIIR) is a membrane protein that plays a central role in the trafficking of lysosomal acid hydrolases into lysosomes via mannose-6-phosphate (M6P) binding domains. In order to maintain cellular metabolic/catabolic homeostasis, newly synthesized lysosomal acid hydrolases are required to bind to M6PR for transit. Acid hydrolases secreted by cells can also be internalized via M6PR residing on the cell membrane and are transported to the lysosomes, a feature that enables enzyme replacement therapy for the treatment of several lysosomal storage disorders. Therefore, a thorough characterization of this receptor is critical to the development of lysosomal enzyme-based therapeutics that utilize M6PR for drug delivery to the lysosome. However, the extracellular domain (ECD) of M6PR is highly complex, containing 15-mannose receptor homology (MRH) domains. In addition, homodimerization of the receptor can occur at the membrane, making its characterization challenging. In this study, a novel human M6PR (hM6PR)-overexpressing cell line originally established for hM6PR cellular uptake assay was utilized for production of hM6PR-ECD, and a novel small molecule biomimetic (aminophenyl-M6P) affinity resin was developed for the purification of M6PR-ECD. The affinity-purified hM6PR-ECD was monomeric, contained 14 intact MRH domains (1-14) and a partial MRH domain 15, and was successfully employed in ELISA-based and surface plasmon resonance-based binding assays to demonstrate its ligand-binding functionality, making it suitable for the evaluation of biotherapeutics that utilize M6PR for cellular internalization.


Subject(s)
Aminophenols/chemistry , Biomimetic Materials/chemistry , Cell Membrane/enzymology , Mannosephosphates/chemistry , Receptor, IGF Type 2/isolation & purification , Amino Acid Sequence , Aminophenols/metabolism , Biomimetic Materials/metabolism , Cell Line, Tumor , Cell Membrane/chemistry , Chromatography, Affinity , Enzyme Assays , Enzyme-Linked Immunosorbent Assay , Fibroblasts/chemistry , Fibroblasts/enzymology , Gene Expression , Humans , Kinetics , Mannosephosphates/metabolism , Protein Domains , Receptor, IGF Type 2/genetics , Receptor, IGF Type 2/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Sequence Alignment , Surface Plasmon Resonance
20.
Article in English | MEDLINE | ID: mdl-31405854

ABSTRACT

Chagas' disease, which is caused by the Trypanosoma cruzi parasite, has become a global health problem that is currently treated with poorly tolerated drugs that require prolonged dosing. Therefore, there is a clinical need for new therapeutic agents that can mitigate these issues. The phosphomannomutase (PMM) and GDP-mannose pyrophosphorylase (GDP-MP) enzymes form part of the de novo biosynthetic pathway to the nucleotide sugar GDP-mannose. This nucleotide sugar is used either directly, or indirectly via the formation of dolichol-phosphomannose, for the assembly of all mannose-containing glycoconjugates. In T. cruzi, mannose-containing glycoconjugates include the cell-surface glycoinositol-phospholipids and the glycosylphosphatidylinositol-anchored mucin-like glycoproteins that dominate the cell surface architectures of all life cycle stages. This makes PMM and GDP-MP potentially attractive targets for a drug discovery program against Chagas' disease. To assess the ligandability of these enzymes in T. cruzi, we have screened 18,117 structurally diverse compounds exploring drug-like chemical space and 16,845 small polar fragment compounds using an assay interrogating the activities of both PMM and GDP-MP enzymes simultaneously. This resulted in 48 small fragment hits, and on retesting 20 were found to be active against the enzymes. Deconvolution revealed that these were all inhibitors of T. cruzi GDP-MP, with compounds 2 and 3 acting as uncompetitive and competitive inhibitors, respectively. Based on these findings, the T. cruzi PMM and GDP-MP enzymes were deemed not ligandable and poorly ligandable, respectively, using small molecules from conventional drug discovery chemical space. This presents a significant hurdle to exploiting these enzymes as therapeutic targets for Chagas' disease.


Subject(s)
Antiprotozoal Agents/pharmacology , Mannose/metabolism , Nucleotidyltransferases/metabolism , Phosphotransferases (Phosphomutases)/metabolism , Trypanosoma cruzi/enzymology , Chagas Disease/parasitology , Drug Discovery/methods , Mannosephosphates/metabolism , Nucleotidyltransferases/genetics , Phosphotransferases (Phosphomutases)/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...