Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.173
Filter
1.
Biotechnol J ; 19(5): e2400098, 2024 May.
Article in English | MEDLINE | ID: mdl-38797728

ABSTRACT

Human carboxypeptidase B1 (hCPB1) is vital for recombinant insulin production, holding substantial value in the pharmaceutical industry. Current challenges include limited hCPB1 enzyme activity. In this study, recombinant hCPB1 efficient expression in Pichia pastoris was achieved. To enhance hCPB1 secretion, we conducted signal peptides screening and deleted the Vps10 sortilin domain, reducing vacuolar mis-sorting. Overexpression of Sec4p increased the fusion of secretory vesicles with the plasma membrane and improved hCPB1 secretion by 20%. Rational protein engineering generated twenty-two single-mutation mutants and identified the A178L mutation resulted in a 30% increase in hCPB1 specific activity. However, all combinational mutations that increased specific activities decreased protein expression levels. Therefore, computer-aided global protein design with PROSS was employed for the aim of improving specific activities and preserving good protein expression. Among the six designed mutants, hCPB1-P6 showed a remarkable 114% increase in the catalytic rate constant (kcat), a 137% decrease in the Michaelis constant (Km), and a 490% increase in catalytic efficiency. Most mutations occurred on the surface of hCPB1-P6, with eight sites mutated to proline. In a 5 L fermenter, hCPB1-P6 was produced by the secretion-enhanced P. pastoris chassis to 199.6 ± 20 mg L-1 with a specific activity of 96 ± 0.32 U mg-1, resulting in a total enzyme activity of 19137 ± 1131 U L-1, demonstrating significant potential for industrial applications.


Subject(s)
Carboxypeptidase B , Cell Membrane , Golgi Apparatus , Protein Engineering , Recombinant Proteins , Humans , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Protein Engineering/methods , Carboxypeptidase B/genetics , Carboxypeptidase B/metabolism , Cell Membrane/metabolism , Cell Membrane/genetics , Golgi Apparatus/metabolism , Golgi Apparatus/genetics , Golgi Apparatus/enzymology , Saccharomycetales/genetics , Saccharomycetales/enzymology , Mutation , Pichia/genetics , Pichia/metabolism , Protein Sorting Signals/genetics , Protein Transport
2.
J Biol Chem ; 300(1): 105564, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38103644

ABSTRACT

The polysialyltransferases ST8SIA2 and ST8SIA4 and their product, polysialic acid (polySia), are known to be related to cancers and mental disorders. ST8SIA2 and ST8SIA4 have conserved amino acid (AA) sequence motifs essential for the synthesis of the polySia structures on the neural cell adhesion molecule. To search for a new motif in the polysialyltransferases, we adopted the in silico Individual Meta Random Forest program that can predict disease-related AA substitutions. The Individual Meta Random Forest program predicted a new eight-amino-acids sequence motif consisting of highly pathogenic AA residues, thus designated as the pathogenic (P) motif. A series of alanine point mutation experiments in the pathogenic motif (P motif) showed that most P motif mutants lost the polysialylation activity without changing the proper enzyme expression levels or localization in the Golgi. In addition, we evaluated the enzyme stability of the P motif mutants using newly established calculations of mutation energy, demonstrating that the subtle change of the conformational energy regulates the activity. In the AlphaFold2 model, we found that the P motif was a buried ß-strand underneath the known surface motifs unique to ST8SIA2 and ST8SIA4. Taken together, the P motif is a novel buried ß-strand that regulates the full activity of polysialyltransferases from the inside of the molecule.


Subject(s)
Mutation , Sialyltransferases , Humans , Amino Acid Motifs/genetics , Amino Acid Substitution , Computer Simulation , Golgi Apparatus/enzymology , Golgi Apparatus/metabolism , Neural Cell Adhesion Molecules/chemistry , Neural Cell Adhesion Molecules/metabolism , Point Mutation , Protein Conformation, beta-Strand , Protein Transport , Random Forest , Sialic Acids/metabolism , Sialyltransferases/chemistry , Sialyltransferases/genetics , Sialyltransferases/metabolism
3.
J Biol Chem ; 299(12): 105473, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37979916

ABSTRACT

Vacuolar H+-ATPases (V-ATPases) are highly conserved multisubunit enzymes that maintain the distinct pH of eukaryotic organelles. The integral membrane a-subunit is encoded by tissue- and organelle-specific isoforms, and its cytosolic N-terminal domain (aNT) modulates organelle-specific regulation and targeting of V-ATPases. Organelle membranes have specific phosphatidylinositol phosphate (PIP) lipid enrichment linked to maintenance of organelle pH. In yeast, the aNT domains of the two a-subunit isoforms bind PIP lipids enriched in the organelle membranes where they reside; these interactions affect activity and regulatory properties of the V-ATPases containing each isoform. Humans have four a-subunit isoforms, and we hypothesize that the aNT domains of these isoforms will also bind to specific PIP lipids. The a1 and a2 isoforms of human V-ATPase a-subunits are localized to endolysosomes and Golgi, respectively. We determined that bacterially expressed Hua1NT and Hua2NT bind specifically to endolysosomal PIP lipids PI(3)P and PI(3,5)P2 and Golgi enriched PI(4)P, respectively. Despite the lack of canonical PIP-binding sites, we identified potential binding sites in the HuaNT domains by sequence comparisons and existing subunit structures and models. We found that mutations at a similar location in the distal loops of both HuaNT isoforms compromise binding to their cognate PIP lipids, suggesting that these loops encode PIP specificity of the a-subunit isoforms. These data suggest a mechanism through which PIP lipid binding could stabilize and activate V-ATPases in distinct organelles.


Subject(s)
Phosphatidylinositol Phosphates , Protein Subunits , Vacuolar Proton-Translocating ATPases , Humans , Binding Sites , Endosomes/enzymology , Endosomes/metabolism , Golgi Apparatus/enzymology , Golgi Apparatus/metabolism , Hydrogen-Ion Concentration , Lysosomes/enzymology , Lysosomes/metabolism , Phosphatidylinositol Phosphates/metabolism , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Protein Subunits/chemistry , Protein Subunits/metabolism , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/metabolism , Substrate Specificity , Vacuolar Proton-Translocating ATPases/chemistry , Vacuolar Proton-Translocating ATPases/metabolism , Protein Domains
4.
J Biol Chem ; 299(10): 105192, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37625589

ABSTRACT

Point mutations in leucine-rich repeat kinase 2 (LRRK2) cause Parkinson's disease and augment LRRK2's kinase activity. However, cellular pathways that endogenously enhance LRRK2 kinase function have not been identified. While overexpressed Rab29 draws LRRK2 to Golgi membranes to increase LRRK2 kinase activity, there is little evidence that endogenous Rab29 performs this function under physiological conditions. Here, we identify Rab38 as a novel physiologic regulator of LRRK2 in melanocytes. In mouse melanocytes, which express high levels of Rab38, Rab32, and Rab29, knockdown (or CRISPR knockout) of Rab38, but not Rab32 or Rab29, decreases phosphorylation of multiple LRRK2 substrates, including Rab10 and Rab12, by both endogenous LRRK2 and exogenous Parkinson's disease-mutant LRRK2. In B16-F10 mouse melanoma cells, Rab38 drives LRRK2 membrane association and overexpressed kinase-active LRRK2 shows striking pericentriolar recruitment, which is dependent on the presence of endogenous Rab38 but not Rab32 or Rab29. Consistently, knockdown or mutation of BLOC-3, the guanine nucleotide exchange factor for Rab38 and Rab32, inhibits Rab38's regulation of LRRK2. Deletion or mutation of LRRK2's Rab38-binding site in the N-terminal armadillo domain decreases LRRK2 membrane association, pericentriolar recruitment, and ability to phosphorylate Rab10. In sum, our data identify Rab38 as a physiologic regulator of LRRK2 function and lend support to a model in which LRRK2 plays a central role in Rab GTPase coordination of vesicular trafficking.


Subject(s)
Intracellular Membranes , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Melanocytes , rab GTP-Binding Proteins , Animals , Mice , Golgi Apparatus/enzymology , Golgi Apparatus/genetics , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Melanocytes/metabolism , Mutation , Parkinson Disease/metabolism , Phosphorylation , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism , HEK293 Cells , Humans , Gene Expression , Protein Domains , Protein Binding , Intracellular Membranes/metabolism
5.
J Mol Biol ; 435(15): 168171, 2023 08 01.
Article in English | MEDLINE | ID: mdl-37285900

ABSTRACT

Carboxypeptidase E (CPE), an essential enzyme in the biosynthetic production line of most peptide hormones and neuropeptides, is predominantly expressed in endocrine tissues and in the nervous system. CPE is active in acidic environments where it cleaves the C'-terminal basic residues of peptide precursors to generate their bioactive form. Consequently, this highly conserved enzyme regulates numerous fundamental biological processes. Here, we combined live-cell microscopy and molecular analysis to examine the intracellular distribution and secretion dynamics of fluorescently tagged CPE. We show that, in non-endocrine cells, tagged-CPE is a soluble luminal protein that is efficiently exported from the ER via the Golgi apparatus to lysosomes. The C'-terminal conserved amphipathic helix serves as a lysosomal and secretory granule targeting and a secretion motif. Following secretion, CPE may be reinternalized into the lysosomes of neighboring cells.


Subject(s)
Carboxypeptidase H , Lysosomes , Carboxypeptidase H/genetics , Carboxypeptidase H/metabolism , Golgi Apparatus/enzymology , Lysosomes/enzymology , Neuropeptides/metabolism
6.
Biochim Biophys Acta Gen Subj ; 1867(9): 130412, 2023 09.
Article in English | MEDLINE | ID: mdl-37348823

ABSTRACT

The remarkable structural diversity of glycans that is exposed at the cell surface and generated along the secretory pathway is tightly regulated by several factors. The recent identification of human glycosylation diseases related to metal transporter defects opened a completely new field of investigation, referred to herein as "metalloglycobiology", on how metal changes can affect the glycosylation and hence the glycan structures that are produced. Although this field is in its infancy, this review aims to go through the different glycosylation steps/pathways that are metal dependent and that could be impacted by metal homeostasis dysregulations.


Subject(s)
Glycomics , Glycosylation , Metals , Polysaccharides , Humans , Cation Transport Proteins/metabolism , Congenital Disorders of Glycosylation/metabolism , Endoplasmic Reticulum/enzymology , Endoplasmic Reticulum/metabolism , Glycomics/trends , Golgi Apparatus/enzymology , Golgi Apparatus/metabolism , Homeostasis , Magnesium/chemistry , Magnesium/metabolism , Metals/chemistry , Metals/metabolism , Oxidation-Reduction , Polysaccharides/chemistry , Polysaccharides/metabolism , Zinc/chemistry , Zinc/metabolism
7.
J Biol Chem ; 298(9): 102281, 2022 09.
Article in English | MEDLINE | ID: mdl-35863437

ABSTRACT

Rab22 and Rab31 belong to the Rab5 subfamily of GTPases that regulates endocytic traffic and endosomal sorting. Rab22 and Rab31 (a.k.a. Rab22b) are closely related and share 87% amino acid sequence similarity, but they show distinct intracellular localization and function in the cell. Rab22 is localized to early endosomes and regulates early endosomal recycling, while Rab31 is mostly localized to the Golgi complex with only a small fraction in the endosomes at steady state. The specific determinants that affect this differential localization, however, are unclear. In this study, we identify a novel membrane targeting domain (MTD) consisting of the C-terminal hypervariable domain (HVD), interswitch loop (ISL), and N-terminal domain as a major determinant of endosomal localization for Rab22 and Rab31, as well as Rab5. Rab22 and Rab31 share the same N-terminal domain, but we find Rab22 chimeras with Rab31 HVD exhibit phenotypic Rab31 localization to the Golgi complex, while Rab31 chimeras with the Rab22 HVD localize to early endosomes, similar to wildtype Rab22. We also find that the Rab22 HVD favors interaction with the early endosomal effector protein Rabenosyn-5, which may stabilize the Rab localization to the endosomes. The importance of effector interaction in endosomal localization is further demonstrated by the disruption of Rab22 endosomal localization in Rabenosyn-5 knockout cells and by the shift of Rab31 to the endosomes in Rabenosyn-5-overexpressing cells. Taken together, we have identified a novel MTD that mediates localization of Rab5 subfamily members to early endosomes via interaction with an effector such as Rabenosyn-5.


Subject(s)
Endosomes , Golgi Apparatus , rab GTP-Binding Proteins , Animals , Cricetinae , Endosomes/enzymology , Golgi Apparatus/enzymology , HEK293 Cells , Humans , PC12 Cells , Protein Domains , Protein Transport , Rats , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Transport Vesicles/metabolism , rab GTP-Binding Proteins/chemistry , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism , rab5 GTP-Binding Proteins/metabolism
8.
ACS Chem Biol ; 17(1): 240-251, 2022 01 21.
Article in English | MEDLINE | ID: mdl-35000377

ABSTRACT

Many cellular processes are dependent on correct pH levels, and this is especially important for the secretory pathway. Defects in pH homeostasis in distinct organelles cause a wide range of diseases, including disorders of glycosylation and lysosomal storage diseases. Ratiometric imaging of the pH-sensitive mutant of green fluorescent protein, pHLuorin, has allowed for targeted pH measurements in various organelles, but the required sequential image acquisition is intrinsically slow and therefore the temporal resolution is unsuitable to follow the rapid transit of cargo between organelles. Therefore, we applied fluorescence lifetime imaging microscopy (FLIM) to measure intraorganellar pH with just a single excitation wavelength. We first validated this method by confirming the pH in multiple compartments along the secretory pathway and compared the pH values obtained by the FLIM-based measurements with those obtained by conventional ratiometric imaging. Then, we analyzed the dynamic pH changes within cells treated with Bafilomycin A1, to block the vesicular ATPase, and Brefeldin A, to block endoplasmic reticulum (ER)-Golgi trafficking. Finally, we followed the pH changes of newly synthesized molecules of the inflammatory cytokine tumor necrosis factor-α while they were in transit from the ER via the Golgi to the plasma membrane. The toolbox we present here can be applied to measure intracellular pH with high spatial and temporal resolution and can be used to assess organellar pH in disease models.


Subject(s)
Hydrogen-Ion Concentration , Optical Imaging/methods , Secretory Pathway , Adenosine Triphosphatases/antagonists & inhibitors , Brefeldin A/pharmacology , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/enzymology , Endoplasmic Reticulum/metabolism , Golgi Apparatus/drug effects , Golgi Apparatus/enzymology , Golgi Apparatus/metabolism , Humans , Macrolides/pharmacology , Microscopy, Fluorescence/methods , Protein Transport
9.
J Biol Chem ; 296: 100315, 2021.
Article in English | MEDLINE | ID: mdl-33485966

ABSTRACT

Lipid flipping in the membrane bilayers is a widespread eukaryotic phenomenon that is catalyzed by assorted P4-ATPases. Its occurrence, mechanism, and importance in apicomplexan parasites have remained elusive, however. Here we show that Toxoplasma gondii, an obligate intracellular parasite with high clinical relevance, can salvage phosphatidylserine (PtdSer) and phosphatidylethanolamine (PtdEtn) but not phosphatidylcholine (PtdCho) probes from its milieu. Consistently, the drug analogs of PtdCho are broadly ineffective in the parasite culture. NBD-PtdSer imported to the parasite interior is decarboxylated to NBD-PtdEtn, while the latter is not methylated to yield PtdCho, which confirms the expression of PtdSer decarboxylase but a lack of PtdEtn methyltransferase activity and suggests a role of exogenous lipids in membrane biogenesis of T. gondii. Flow cytometric quantitation of NBD-probes endorsed the selectivity of phospholipid transport and revealed a dependence of the process on energy and protein. Accordingly, our further work identified five P4-ATPases (TgP4-ATPase1-5), all of which harbor the signature residues and motifs required for phospholipid flipping. Of the four proteins expressed during the lytic cycle, TgP4-ATPase1 is present in the apical plasmalemma; TgP4-ATPase3 resides in the Golgi network along with its noncatalytic partner Ligand Effector Module 3 (TgLem3), whereas TgP4-ATPase2 and TgP4-ATPase5 localize in the plasmalemma as well as endo/cytomembranes. Last but not least, auxin-induced degradation of TgP4-ATPase1-3 impaired the parasite growth in human host cells, disclosing their crucial roles during acute infection. In conclusion, we show selective translocation of PtdEtn and PtdSer at the parasite surface and provide the underlying mechanistic and physiological insights in a model eukaryotic pathogen.


Subject(s)
Adenosine Triphosphatases/genetics , Lipid Bilayers/metabolism , Toxoplasma/genetics , Toxoplasmosis/genetics , Adenosine Triphosphatases/chemistry , Cell Membrane/genetics , Cell Membrane/metabolism , Flow Cytometry , Glycerophospholipids/metabolism , Golgi Apparatus/chemistry , Golgi Apparatus/enzymology , Humans , Lipid Bilayers/chemistry , Lipids/chemistry , Lipids/genetics , Phosphatidylcholines/genetics , Phosphatidylcholines/metabolism , Phosphatidylethanolamines/genetics , Phosphatidylethanolamines/metabolism , Phosphatidylserines/metabolism , Toxoplasma/enzymology , Toxoplasma/pathogenicity , Toxoplasmosis/parasitology
10.
Carbohydr Polym ; 255: 117477, 2021 Mar 01.
Article in English | MEDLINE | ID: mdl-33436240

ABSTRACT

The cell surface and extracellular matrix polysaccharide, heparan sulfate (HS) conveys chemical information to control crucial biological processes. HS chains are synthesized in a non-template driven process mainly in the Golgi apparatus, involving a large number of enzymes capable of subtly modifying its substitution pattern, hence, its interactions and biological effects. Changes in the localization of HS-modifying enzymes throughout the Golgi were found to correlate with changes in the structure of HS, rather than protein expression levels. Following BFA treatment, the HS-modifying enzymes localized preferentially in COPII vesicles and at the trans-Golgi. Shortly after heparin treatment, the HS-modifying enzyme moved from cis to trans-Golgi, which coincided with increased HS sulfation. Finally, it was shown that COPI subunits and Sec24 gene expression changed. Collectively, these findings demonstrate that knowledge of the ER-Golgi dynamics of HS-modifying enzymes via vesicular trafficking is a critical prerequisite for the complete delineation of HS biosynthesis.


Subject(s)
COP-Coated Vesicles/enzymology , Endoplasmic Reticulum/enzymology , Golgi Apparatus/enzymology , Heparitin Sulfate/biosynthesis , Biological Transport/drug effects , Brefeldin A/pharmacology , COP-Coated Vesicles/genetics , Cell Membrane/chemistry , Cell Membrane/drug effects , Cell Membrane/enzymology , Endoplasmic Reticulum/chemistry , Endoplasmic Reticulum/drug effects , Gene Expression Regulation , Golgi Apparatus/chemistry , Golgi Apparatus/drug effects , Heparin/pharmacology , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/enzymology , Humans , Plasmids/chemistry , Plasmids/metabolism , Primary Cell Culture , Transfection , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism
11.
Cells ; 11(1)2021 12 29.
Article in English | MEDLINE | ID: mdl-35011665

ABSTRACT

Mutations in the apically located kidney Na-K-2Cl cotransporter NKCC2 cause type I Bartter syndrome, a life-threatening kidney disorder. We previously showed that transport from the ER represents the limiting phase in NKCC2 journey to the cell surface. Yet very little is known about the ER quality control components specific to NKCC2 and its disease-causing mutants. Here, we report the identification of Golgi alpha1, 2-mannosidase IA (ManIA) as a novel binding partner of the immature form of NKCC2. ManIA interaction with NKCC2 takes place mainly at the cis-Golgi network. ManIA coexpression decreased total NKCC2 protein abundance whereas ManIA knock-down produced the opposite effect. Importantly, ManIA coexpression had a more profound effect on NKCC2 folding mutants. Cycloheximide chase assay showed that in cells overexpressing ManIA, NKCC2 stability and maturation are heavily hampered. Deleting the cytoplasmic region of ManIA attenuated its interaction with NKCC2 and inhibited its effect on the maturation of the cotransporter. ManIA-induced reductions in NKCC2 expression were offset by the proteasome inhibitor MG132. Likewise, kifunensine treatment greatly reduced ManIA effect, strongly suggesting that mannose trimming is involved in the enhanced ERAD of the cotransporter. Moreover, depriving ManIA of its catalytic domain fully abolished its effect on NKCC2. In summary, our data demonstrate the presence of a ManIA-mediated ERAD pathway in renal cells promoting retention and degradation of misfolded NKCC2 proteins. They suggest a model whereby Golgi ManIA contributes to ERAD of NKCC2, by promoting the retention, recycling, and ERAD of misfolded proteins that initially escape protein quality control surveillance within the ER.


Subject(s)
Endoplasmic Reticulum-Associated Degradation , Golgi Apparatus/enzymology , Mannosidases/metabolism , Solute Carrier Family 12, Member 1/metabolism , Animals , Cell Line , Humans , Mannose/metabolism , Mannosidases/chemistry , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Opossums , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Protein Domains , Protein Folding , Protein Stability
12.
Chem Pharm Bull (Tokyo) ; 68(8): 753-761, 2020.
Article in English | MEDLINE | ID: mdl-32741916

ABSTRACT

The genes GLB1 and GALC encode GLB1 isoform 1 and galactocerebrosidase, respectively, which exhibit ß-galactosidase activity in human lysosomes. GLB1 isoform 1 has been reported to play roles in rare lysosomal storage diseases. Further, its ß-galactosidase activity is the most widely used biomarker of senescent and aging cells; hence, it is called senescence-associated ß-galactosidase. Galactocerebrosidase plays roles in Krabbe disease. We previously reported a novel ß-galactosidase activity in the Golgi apparatus of human cells; however, the protein responsible for this activity could not be identified. Inhibitor-derived chemical probes can serve as powerful tools to identify the responsible protein. In this study, we first constructed a cell-based high-throughput screening (HTS) system for Golgi ß-galactosidase inhibitors, and then screened inhibitors from two compound libraries using the HTS system, in vitro assay, and cytotoxicity assay. An isoflavone derivative was identified among the final Golgi ß-galactosidase inhibitor compound hits. Molecular docking simulations were performed to redesign the isoflavone derivative into a more potent inhibitor, and six designed derivatives were then synthesized. One of the derivatives, ARM07, exhibited potent inhibitory activity against ß-galactosidase, with an IC50 value of 14.8 µM and competitive inhibition with Ki value of 13.3 µM. Furthermore, the in vitro and cellular inhibitory activities of ARM07 exceeded those of deoxygalactonojirimycin. ARM07 may contribute to the development of affinity-based chemical probes to identify the protein responsible for the newly discovered Golgi ß-galactosidase activity. The therapeutic relevance of ARM07 against lysosomal storage diseases and its effect on senescent cells should be evaluated further.


Subject(s)
Enzyme Inhibitors/chemical synthesis , Golgi Apparatus/enzymology , Isoflavones/chemistry , beta-Galactosidase/antagonists & inhibitors , Binding Sites , Cell Line , Crystallography, X-Ray , Drug Design , Enzyme Inhibitors/metabolism , High-Throughput Screening Assays , Humans , Inhibitory Concentration 50 , Isoflavones/metabolism , Kinetics , Molecular Docking Simulation , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Structure-Activity Relationship , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
13.
J Cell Biol ; 219(8)2020 08 03.
Article in English | MEDLINE | ID: mdl-32479594

ABSTRACT

Matrix metalloproteinases (MMPs) degrade several ECM components and are crucial modulators of cell invasion and tissue organization. Although much has been reported about their function in remodeling ECM in health and disease, their trafficking across the Golgi apparatus remains poorly understood. Here we report that the cis-Golgi protein nucleobindin-1 (NUCB1) is critical for MMP2 and MT1-MMP trafficking along the Golgi apparatus. This process is Ca2+-dependent and is required for invasive MDA-MB-231 cell migration as well as for gelatin degradation in primary human macrophages. Our findings emphasize the importance of NUCB1 as an essential component of MMP transport and its overall impact on ECM remodeling.


Subject(s)
Breast Neoplasms/enzymology , Extracellular Matrix/enzymology , Golgi Apparatus/enzymology , Macrophages/enzymology , Matrix Metalloproteinase 14/metabolism , Matrix Metalloproteinase 2/metabolism , Nucleobindins/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Calcium/metabolism , Calcium Signaling , Cell Movement , Extracellular Matrix/pathology , Female , Gelatin/metabolism , HEK293 Cells , HeLa Cells , Humans , Matrix Metalloproteinase 14/genetics , Matrix Metalloproteinase 2/genetics , Nucleobindins/genetics , Protein Transport , Proteolysis , Time Factors
14.
Elife ; 92020 06 29.
Article in English | MEDLINE | ID: mdl-32597757

ABSTRACT

The synthesis of eukaryotic glycans - branched sugar oligomers attached to cell-surface proteins and lipids - is organized like a factory assembly line. Specific enzymes within successive compartments of the Golgi apparatus determine where new monomer building blocks are linked to the growing oligomer. These enzymes act promiscuously and stochastically, causing microheterogeneity (molecule-to-molecule variability) in the final oligomer products. However, this variability is tightly controlled: a given eukaryotic protein type is typically associated with a narrow, specific glycan oligomer profile. Here, we use ideas from the mathematical theory of self-assembly to enumerate the enzymatic causes of oligomer variability and show how to eliminate each cause. We rigorously demonstrate that cells can specifically synthesize a larger repertoire of glycan oligomers by partitioning promiscuous enzymes across multiple Golgi compartments. This places limits on biomolecular assembly: glycan microheterogeneity becomes unavoidable when the number of compartments is limited, or enzymes are excessively promiscuous.


Subject(s)
Golgi Apparatus/metabolism , Eukaryota/enzymology , Eukaryota/metabolism , Glycogen Debranching Enzyme System/metabolism , Golgi Apparatus/enzymology , Multiprotein Complexes/metabolism , Polysaccharides/metabolism , Protein Multimerization , Stochastic Processes
15.
J Clin Invest ; 130(8): 4118-4132, 2020 08 03.
Article in English | MEDLINE | ID: mdl-32597833

ABSTRACT

Lysosomal enzymes are synthesized in the endoplasmic reticulum (ER) and transferred to the Golgi complex by interaction with the Batten disease protein CLN8 (ceroid lipofuscinosis, neuronal, 8). Here we investigated the relationship of this pathway with CLN6, an ER-associated protein of unknown function that is defective in a different Batten disease subtype. Experiments focused on protein interaction and trafficking identified CLN6 as an obligate component of a CLN6-CLN8 complex (herein referred to as EGRESS: ER-to-Golgi relaying of enzymes of the lysosomal system), which recruits lysosomal enzymes at the ER to promote their Golgi transfer. Mutagenesis experiments showed that the second luminal loop of CLN6 is required for the interaction of CLN6 with the enzymes but dispensable for interaction with CLN8. In vitro and in vivo studies showed that CLN6 deficiency results in inefficient ER export of lysosomal enzymes and diminished levels of the enzymes at the lysosome. Mice lacking both CLN6 and CLN8 did not display aggravated pathology compared with the single deficiencies, indicating that the EGRESS complex works as a functional unit. These results identify CLN6 and the EGRESS complex as key players in lysosome biogenesis and shed light on the molecular etiology of Batten disease caused by defects in CLN6.


Subject(s)
Endoplasmic Reticulum/enzymology , Golgi Apparatus/enzymology , Lysosomes/enzymology , Membrane Proteins/metabolism , Multiprotein Complexes/metabolism , Animals , Endoplasmic Reticulum/genetics , Golgi Apparatus/genetics , Lysosomes/genetics , Membrane Proteins/genetics , Mice , Mice, Knockout , Multiprotein Complexes/genetics , Neuronal Ceroid-Lipofuscinoses/enzymology , Neuronal Ceroid-Lipofuscinoses/genetics , Neuronal Ceroid-Lipofuscinoses/pathology , Protein Transport/genetics
16.
Biochem Soc Trans ; 48(3): 1129-1138, 2020 06 30.
Article in English | MEDLINE | ID: mdl-32573677

ABSTRACT

Glycosylphosphatidylinositol (GPI) is a glycolipid added to the C-terminus of a large variety of proteins in eukaryotes, thereby anchoring these proteins to the cell surface. More than 150 different human proteins are modified with GPI, and GPI-anchored proteins (GPI-APs) play critical roles in embryogenesis, neurogenesis, immunity, and fertilization. GPI-APs are biosynthesized in the endoplasmic reticulum (ER) and transported to the plasma membrane via the Golgi apparatus. During transport, GPI-APs undergo structural remodeling that is important for the efficient folding and sorting of GPI-APs. Asparagine-linked glycan-dependent folding and deacylation by PGAP1 work together to ensure that correctly folded GPI-APs are transported from the ER to the Golgi. Remodeling of the GPI lipid moiety is critical for the association of GPI-APs with lipid rafts. On the cell surface, certain GPI-APs are cleaved by GPI cleavage enzymes and released from the membrane, a key event in processes such as spermatogenesis and neurogenesis. In this review, we discuss the enzymes involved in GPI-AP biosynthesis and the fate of GPI-APs in mammalian cells, with a focus on the assembly, folding, degradation, and cleavage of GPI-APs.


Subject(s)
Cell Membrane/metabolism , Endoplasmic Reticulum/enzymology , Glycosylphosphatidylinositols/biosynthesis , Golgi Apparatus/enzymology , Animals , Humans , Male , Membrane Microdomains/enzymology , Membrane Proteins/metabolism , Neurogenesis , Protein Domains , Protein Folding , Protein Transport , Spermatogenesis
17.
mBio ; 11(3)2020 06 02.
Article in English | MEDLINE | ID: mdl-32487759

ABSTRACT

Many species of pathogenic fungi deploy the unfolded protein response (UPR) to expand the folding capacity of the endoplasmic reticulum (ER) in proportion to the demand for virulence-related proteins that traffic through the secretory pathway. Although Ca2+ plays a pivotal role in ER function, the mechanism by which transcriptional upregulation of the protein folding machinery is coordinated with Ca2+ homeostasis is incompletely understood. In this study, we investigated the link between the UPR and genes encoding P-type Ca2+-ATPases in the human-pathogenic mold Aspergillus fumigatus We demonstrate that acute ER stress increases transcription of the srcA gene, encoding a member of the sarco/endoplasmic reticulum Ca2+-ATPase (SERCA) family, as well as that of pmrA, encoding a secretory pathway Ca2+-ATPase (SPCA) in the Golgi membrane. Loss of the UPR transcription factor HacA prevented the induction of srcA and pmrA transcription during ER stress, defining these ER/Golgi Ca2+ pumps as novel downstream targets of this pathway. While deletion of srcA alone caused no major deficiencies, a ΔsrcA/ΔpmrA mutant displayed a severe polarity defect, was hypersensitive to ER stress, and showed attenuated virulence. In addition, cell wall analyses revealed a striking reduction in mannose levels in the absence of both Ca2+ pumps. The ΔhacA mutant was hypersensitive to agents that block calcineurin-dependent signaling, consistent with a functional coupling between the UPR and Ca2+ homeostasis. Together, these findings demonstrate that the UPR integrates the need for increased levels of chaperone and folding enzymes with an influx of Ca2+ into the secretory pathway to support fungal growth, stress adaptation, and pathogenicity.IMPORTANCE The UPR is an intracellular signal transduction pathway that maintains homeostasis of the ER. The pathway is also tightly linked to the expression of virulence-related traits in diverse species of human-pathogenic and plant-pathogenic fungal species, including the predominant mold pathogen infecting humans, Aspergillus fumigatus Despite advances in the understanding of UPR signaling, the linkages and networks that are governed by this pathway are not well defined. In this study, we revealed that the UPR is a major driving force for stimulating Ca2+ influx at the ER and Golgi membranes and that the coupling between the UPR and Ca2+ import is important for virulence, cell wall biosynthesis, and resistance to antifungal compounds that inhibit Ca2+ signaling.


Subject(s)
Adenosine Triphosphatases/metabolism , Aspergillus fumigatus/enzymology , Aspergillus fumigatus/pathogenicity , Cell Wall/physiology , Endoplasmic Reticulum Stress , Unfolded Protein Response , A549 Cells , Alveolar Epithelial Cells/microbiology , Animals , Aspergillus fumigatus/genetics , Calcium/metabolism , Endoplasmic Reticulum/enzymology , Female , Fungal Proteins/genetics , Fungal Proteins/metabolism , Golgi Apparatus/enzymology , Humans , Male , Mice , Signal Transduction , Virulence
18.
Cell Adh Migr ; 14(1): 96-105, 2020 12.
Article in English | MEDLINE | ID: mdl-32419574

ABSTRACT

CD147 is a widely expressed matrix metalloproteinase inducer involved in the regulation of cell migration. The high glycosylation and ability to undergo oligomerization have been linked to CD147 function, yet there is limited understanding on the molecular mechanisms behind these processes. The current study demonstrates that the expression of Golgi α1,2-mannosidase I is key to maintaining the cell surface organization of CD147 during cell migration. Using an in vitro model of stratified human corneal epithelial wound healing, we show that CD147 is clustered within lateral plasma membranes at the leading edge of adjacent migrating cells. This localization correlates with a surge in matrix metalloproteinase activity and an increase in the expression of α1,2-mannosidase subtype IC (MAN1C1). Global inhibition of α1,2-mannosidase I activity with deoxymannojirimycin markedly attenuates the glycosylation of CD147 and disrupts its surface distribution at the leading edge, concomitantly reducing the expression of matrix metalloproteinase-9. Likewise, treatment with deoxymannojirimycin or siRNA-mediated knockdown of MAN1C1 impairs the ability of the carbohydrate-binding protein galectin-3 to stimulate CD147 clustering in unwounded cells. We conclude that the mannose-trimming activity of α1,2-mannosidase I coordinates the clustering and compartmentalization of CD147 that follows an epithelial injury.


Subject(s)
Basigin/metabolism , Cell Movement , Epithelial Cells/cytology , Epithelial Cells/metabolism , Golgi Apparatus/enzymology , Mannosidases/metabolism , Cell Membrane/metabolism , Epithelium, Corneal/cytology , Galectin 3/metabolism , Humans , Polysaccharides/chemistry , Polysaccharides/metabolism
19.
Bioorg Med Chem ; 28(11): 115492, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32291147

ABSTRACT

Effective chemotherapy for solid cancers is challenging due to a limitation in permeation that prevents anticancer drugs from reaching the center of the tumor, therefore unable to limit cancer cell growth. To circumvent this issue, we planned to apply the drugs directly at the center by first collapsing the outer structure. For this, we focused on cell-cell communication (CCC) between N-glycans and proteins at the tumor cell surface. Mature N-glycans establish CCC; however, CCC is hindered when numerous immature N-glycans are present at the cell surface. Inhibition of Golgi mannosidases (GMs) results in the transport of immature N-glycans to the cell surface. This can be employed to disrupt CCC. Here, we describe the molecular design and synthesis of an improved GM inhibitor with a non-sugar mimic scaffold that was screened from a compound library. The synthesized compounds were tested for enzyme inhibition ability and inhibition of spheroid formation using cell-based methods. Most of the compounds designed and synthesized exhibited GM inhibition at the cellular level. Of those, AR524 had higher inhibitory activity than a known GM inhibitor, kifunensine. Moreover, AR524 inhibited spheroid formation of human malignant cells at low concentration (10 µM), based on the disruption of CCC by GM inhibition.


Subject(s)
Drug Design , Enzyme Inhibitors/pharmacology , Golgi Apparatus/enzymology , Mannosidases/antagonists & inhibitors , Spheroids, Cellular/drug effects , Cell Line, Tumor , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Mannosidases/metabolism , Molecular Structure , Optical Imaging , Spheroids, Cellular/metabolism , Structure-Activity Relationship
20.
Exp Cell Res ; 390(2): 111961, 2020 05 15.
Article in English | MEDLINE | ID: mdl-32209306

ABSTRACT

N-alpha-acetyltransferase 80 (NAA80) was recently demonstrated to acetylate the N-terminus of actin, with NAA80 knockout cells showing actin cytoskeleton-related phenotypes, such as increased formation of membrane protrusions and accelerated migration. Here we report that NAA80 knockout cells additionally display fragmentation of the Golgi apparatus. We further employed rescue assays to demonstrate that this phenotype is connected to the ability of NAA80 to modify actin. Thus, re-expression of NAA80, which leads to re-establishment of actin's N-terminal acetyl group, rescued the Golgi fragmentation, whereas a catalytic dead NAA80 mutant could neither restore actin Nt-acetylation nor Golgi structure. The Golgi phenotype of NAA80 KO cells was shared by both migrating and non-migrating cells and live-cell imaging indicated increased Golgi dynamics in migrating NAA80 KO cells. Finally, we detected a drastic increase in the amount of F-actin in cells lacking NAA80, suggesting a causal relationship between this effect and the observed re-organization of Golgi structure. The findings further underscore the importance of actin Nt-acetylation and provide novel insight into its cellular roles, suggesting a mechanistic link between actin modification state and Golgi organization.


Subject(s)
Acetyltransferases/genetics , Actin Cytoskeleton/enzymology , Actins/genetics , Actins/metabolism , Golgi Apparatus/enzymology , Protein Processing, Post-Translational , Acetylation , Acetyltransferases/deficiency , Actin Cytoskeleton/ultrastructure , Cell Differentiation , Cell Line, Tumor , Cell Movement , Fibroblasts/metabolism , Fibroblasts/ultrastructure , Golgi Apparatus/ultrastructure , Humans , Phenotype , Time-Lapse Imaging
SELECTION OF CITATIONS
SEARCH DETAIL
...