Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters











Publication year range
1.
Methods Mol Biol ; 1321: 73-90, 2015.
Article in English | MEDLINE | ID: mdl-26082216

ABSTRACT

While yeast are lower eukaryotic organisms, they share many common features and biological processes with higher eukaryotes. As such, yeasts have been used as model organisms to facilitate our understanding of such features and processes. To this end, a large number of powerful genetic tools have been developed to investigate and manipulate these organisms. Going hand-in-hand with these genetic tools is the ability to efficiently scale up the fermentation of these organisms, thus making them attractive hosts for the production of recombinant proteins. A key feature of producing recombinant proteins in yeast is that these proteins can be readily secreted into the culture supernatant, simplifying any downstream processing. A consequence of this secretion is that the proteins typically pass through the secretory pathway, during which they may be exposed to various posttranslational modifications. The addition of glycans is one such modification. Unfortunately, while certain aspects of glycosylation are shared between lower and higher eukaryotes, significant differences exist. Over the last two decades much research has focused on engineering the glycosylation pathways of yeast to more closely resemble those of higher eukaryotes, particularly those of humans for the production of therapeutic proteins. In the current review we shall highlight some of the key achievements in yeast glyco-engineering which have led to humanization of both the N- and O-linked glycosylation pathways.


Subject(s)
Fungal Proteins/genetics , Recombinant Proteins/genetics , Yeasts/genetics , Genetic Engineering/methods , Glycosylation , Humans , Polysaccharides/genetics , Protein Processing, Post-Translational/genetics
2.
Appl Microbiol Biotechnol ; 99(9): 3913-27, 2015 May.
Article in English | MEDLINE | ID: mdl-25381909

ABSTRACT

The methylotrophic yeast Pichia pastoris is an attractive expression system due to its ability to secrete large amounts of recombinant protein, with the potential for glycosylation. Advances in glycoengineering of P. pastoris have successfully demonstrated the humanization of both the N- and O-linked glycosylation pathways in this organism. However, in certain cases, the presence of O-linked glycans on a therapeutic protein may not be desirable. Recently, we have reported the in vitro utility of jack bean α-1,2/3/6-mannosidase to remove O-linked mannose from intact undenatured glycoproteins produced in glycoengineered P. pastoris. However, one caveat of this strategy is that jack bean mannosidase has yet to be cloned and as such is only available as crude cellular extracts. This raises several concerns for using this reagent to treat large preparations of therapeutic proteins generated in P. pastoris. Therefore, we postulated that lysosomal mannosidases which have been cloned and demonstrated to have similar activities to jack bean mannosidase on N-linked glycans would also process O-linked glycans in a similar fashion. To this end, we screened a panel of recombinant lysosomal mannosidases from different organisms and identified several which cannot only reduce extended O-linked mannose chains but which can also hydrolyze the Man-α-O-Ser/Thr glycosidic bond on intact glycoproteins. As such, not only do we show for the first time the utility of lysosomal mannosidase for O-linked mannose processing, but since this is a recombinant enzyme, it has several benefits over the use of crude jack bean mannosidase extracts.


Subject(s)
Glycoproteins/metabolism , Lysosomes/enzymology , Mannose/metabolism , Mannosidases/metabolism , Glycoproteins/genetics , Mannosidases/genetics , Pichia/genetics , Pichia/metabolism , Protein Processing, Post-Translational , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
3.
Appl Microbiol Biotechnol ; 98(6): 2573-83, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24526360

ABSTRACT

Yeast are important production platforms for the generation of recombinant proteins. Nonetheless, their use has been restricted in the production of therapeutic proteins due to differences in their glycosylation profile with that of higher eukaryotes. The yeast strain Pichia pastoris is an industrially important organism. Recent advances in the glycoengineering of this strain offer the potential to produce therapeutic glycoproteins with sialylated human-like N- and O-linked glycans. However, like higher eukaryotes, yeast also express numerous proteases, many of which are either localized to the secretory pathway or pass through it en route to their final destination. As a consequence, nondesirable proteolysis of some recombinant proteins may occur, with the specific cleavage being dependent on the class of protease involved. Dipeptidyl aminopeptidases (DPP) are a class of proteolytic enzymes which remove a two-amino acid peptide from the N-terminus of a protein. In P. pastoris, two such enzymes have been identified, Ste13p and Dap2p. In the current report, we demonstrate that while the knockout of STE13 alone may protect certain proteins from N-terminal clipping, other proteins may require the double knockout of both STE13 and DAP2. As such, this understanding of DPP activity enhances the utility of the P. pastoris expression system, thus facilitating the production of recombinant therapeutic proteins with their intact native sequences.


Subject(s)
Biological Products/metabolism , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/genetics , Gene Knockout Techniques , Peptides/metabolism , Pichia/enzymology , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/metabolism , Peptides/genetics , Pichia/genetics , Pichia/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
4.
Appl Microbiol Biotechnol ; 98(6): 2545-54, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24526361

ABSTRACT

The methylotrophic yeast Pichia pastoris is an attractive expression system for heterologous protein production due to its ability to perform posttranslational modifications, such as glycosylation, and secrete large amounts of recombinant protein. However, the structures of N- and O-linked oligosaccharide chains in yeast differ significantly from those of mammalian cells. The most common O-linked glycan structures added by P. pastoris are typically polymers of between one and four α-linked mannose residues, with a subset of glycans being capped by a ß-1,2-mannose disaccharide or phosphomannose residue. Such mannosylation of recombinant proteins is considered a key factor in immunomodulation, with mannose-specific receptors binding and promoting enhanced immune responses. As a result of engineering the N-linked glycosylation pathway of P. pastoris, the recombinant proteins expressed in this system are devoid of phospho- and ß-mannose on O-linked glycans, leaving only α-mannose polymers. Here we screen a library of α-mannosidases for their ability to decrease the extent of O-mannosylation on glycoproteins secreted from this expression system. In doing so, we demonstrate the utility of the α-1,2/3/6-mannosidase from Jack bean in not only reducing extended O-linked mannose chains but also in specifically hydrolyzing the Man-α-O-Ser/Thr glycosidic bond on intact glycoproteins. As such, this presents for the first time a strategy to remove O-linked glycosylation from intact glycoproteins expressed in P. pastoris. We additionally show that this strategy can be used to significantly decrease the extent of O-mannosylation on commercial products produced in other similar expression systems.


Subject(s)
Canavalia/enzymology , Glycoproteins/metabolism , Mannose/metabolism , Mannosidases/metabolism
5.
Glycobiology ; 23(10): 1192-203, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23893788

ABSTRACT

The methylotrophic yeast, Pichia pastoris, is an important organism used for the production of therapeutic proteins. Previously, we have reported the glycoengineering of this organism to produce human-like N-linked glycans but up to now no one has addressed engineering the O-linked glycosylation pathway. Typically, O-linked glycans produced by wild-type P. pastoris are linear chains of four to five α-linked mannose residues, which may be capped with ß- or phospho-mannose. Previous genetic engineering of the N-linked glycosylation pathway of P. pastoris has eliminated both of these two latter modifications, resulting in O-linked glycans which are linear α-linked mannose structures. Here, we describe a method for the co-expression of an α-1,2-mannosidase, which reduces these glycans to primarily a single O-linked mannose residue. In doing so, we have reduced the potential of these glycans to interact with carbohydrate-binding proteins, such as dendritic cell-specific intercellular adhesion molecule-3-grabbing non-integrin. Furthermore, the introduction of the enzyme protein-O-linked-mannose ß-1,2-N-acetylglucosaminyltransferase 1, resulted in the capping of the single O-linked mannose residues with N-acetylglucosamine. Subsequently, this glycoform was extended into human-like sialylated glycans, similar in structure to α-dystroglycan-type glycoforms. As such, this represents the first example of sialylated O-linked glycans being produced in yeast and extends the utility of the P. pastoris production platform beyond N-linked glycosylated biotherapeutics to include molecules possessing O-linked glycans.


Subject(s)
Mannose/biosynthesis , Metabolic Engineering/methods , Pichia/metabolism , alpha-Mannosidase/metabolism , Pichia/growth & development , Protein Engineering , alpha-Mannosidase/genetics
6.
PLoS One ; 8(7): e68325, 2013.
Article in English | MEDLINE | ID: mdl-23840891

ABSTRACT

The methylotrophic yeast, Pichiapastoris, is an important organism used for the production of therapeutic proteins. However, the presence of fungal-like glycans, either N-linked or O-linked, can elicit an immune response or enable the expressed protein to bind to mannose receptors, thus reducing their efficacy. Previously we have reported the elimination of ß-linked glycans in this organism. In the current report we have focused on reducing the O-linked mannose content of proteins produced in P. pastoris, thereby reducing the potential to bind to mannose receptors. The initial step in the synthesis of O-linked glycans in P. pastoris is the transfer of mannose from dolichol-phosphomannose to a target protein in the yeast secretory pathway by members of the protein-O-mannosyltransferase (PMT) family. In this report we identify and characterize the members of the P. pastoris PMT family. Like Candida albicans, P. pastoris has five PMT genes. Based on sequence homology, these PMTs can be grouped into three sub-families, with both PMT1 and PMT2 sub-families possessing two members each (PMT1 and PMT5, and PMT2 and PMT6, respectively). The remaining sub-family, PMT4, has only one member (PMT4). Through gene knockouts we show that PMT1 and PMT2 each play a significant role in O-glycosylation. Both, by gene knockouts and the use of Pmt inhibitors we were able to significantly reduce not only the degree of O-mannosylation, but also the chain-length of these glycans. Taken together, this reduction of O-glycosylation represents an important step forward in developing the P. pastoris platform as a suitable system for the production of therapeutic glycoproteins.


Subject(s)
Mannosyltransferases/genetics , Pichia/enzymology , Pichia/genetics , Candida albicans/enzymology , Candida albicans/genetics , Gene Knockout Techniques , Genes, Fungal , Glycosylation , Mannosyltransferases/metabolism , Phylogeny , Pichia/metabolism , Polysaccharides/metabolism
7.
Pharm Res ; 30(3): 803-12, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23135825

ABSTRACT

PURPOSE: P. pastoris has previously been genetically engineered to generate strains that are capable of producing mammalian-like glycoforms. Our objective was to investigate the correlation between sialic acid content and pharmacokinetic properties of recombinant TNFR2:Fc fusion proteins generated in glycoengineered P. pastoris strains. METHODS: TNFR2:Fc fusion proteins were generated with varying degrees of sialic acid content. The pharmacokinetic properties of these proteins were assessed by intravenous and subcutaneous routes of administration in rats. The binding of these variants to FcRn were also evaluated for possible correlations between in vitro binding and in vivo PK. RESULTS: The pharmacokinetic profiles of recombinant TNFR2:Fc produced in P. pastoris demonstrated a direct positive correlation between the extent of glycoprotein sialylation and in vivo pharmacokinetic properties. Furthermore, recombinant TNFR2:Fc produced in glycoengineered Pichia, with a similar sialic acid content to CHO-produced etanercept, demonstrated similar in vivo pharmacokinetic properties to the commercial material. In vitro surface plasmon resonance FcRn binding at pH6.0 showed an inverse relationship between sialic acid content and receptor binding affinity, with the higher affinity binders having poorer in vivo PK profiles. CONCLUSIONS: Sialic acid content is a critical attribute for modulating the pharmacokinetics of recombinant TNFR2:Fc produced in glycoengineered P. pastoris.


Subject(s)
Immunoglobulin G/blood , Immunoglobulin G/genetics , Immunosuppressive Agents/blood , Pichia/genetics , Receptors, Tumor Necrosis Factor/blood , Receptors, Tumor Necrosis Factor/genetics , Animals , Cloning, Molecular , Etanercept , Genetic Engineering , Glycosylation , Histocompatibility Antigens Class I/metabolism , Humans , Immunoglobulin G/chemistry , Immunoglobulin G/metabolism , Immunosuppressive Agents/chemistry , Immunosuppressive Agents/metabolism , Male , N-Acetylneuraminic Acid/analysis , Protein Binding , Rats , Rats, Sprague-Dawley , Receptors, Fc/metabolism , Receptors, Tumor Necrosis Factor/chemistry , Receptors, Tumor Necrosis Factor/metabolism
8.
J Pharm Sci ; 101(12): 4414-8, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22987365

ABSTRACT

Erythropoietin (EPO) is an important molecule in the erythropoiesis and various forms of EPO have been marketed in managing anemia in humans. Long acting EPOs for less frequent dosing have been generated either by increasing the number of glycosylation sites of the EPO molecule or by linking it to a polyethylene glycol (PEG). We have generated recombinant human EPO (rhEPO) using glycoengineered Pichia pastoris strains and evaluated the pharmacokinetics (PK) in rats of this molecule linked to a 40 kDa PEG (PEGylated rhEPO), in relation to its glycosylation patterns. As expected, the PEGylated rhEPO exhibited a significant improvement in half-life of serum when compared with the non-PEGylated version. Interestingly, the PK properties of the PEGylated rhEPO molecule were also significantly influenced by the glycosylation profile. Specifically, PEGylated rhEPO with a significantly higher sialic acid content in the biantennary structure (high A2) exhibited lower systemic clearance and higher systemic exposure than those with a lower sialic acid content (low A2) following either intravenous or subcutaneous administrations. These results suggest that A2 content may be one of the important criteria for release in manufacturing PEGylated rhEPO to ensure consistent PK.


Subject(s)
Erythropoietin/blood , Erythropoietin/chemistry , Polyethylene Glycols/chemistry , Sialic Acids/chemistry , Animals , Carbohydrate Sequence , Erythropoietin/genetics , Glycosylation , Half-Life , Humans , Male , Molecular Sequence Data , Pichia/genetics , Protein Engineering , Rats , Rats, Sprague-Dawley , Recombinant Proteins/blood , Recombinant Proteins/chemistry , Recombinant Proteins/genetics
9.
Methods Mol Biol ; 899: 315-23, 2012.
Article in English | MEDLINE | ID: mdl-22735962

ABSTRACT

The secretory pathway of the yeast Pichia pastoris has been engineered to produce complex human-type N-glycans (Choi et al., Proc Natl Acad Sci USA 100:5022-5027, 2003; Hamilton et al., Science 301:1244-1246, 2003; Hamilton et al., Science 313:1441-1443, 2006). In contrast to the heterogeneous glycans produced on the therapeutic glycoproteins expressed in mammalian cell lines, glycoengineered P. pastoris can be designed to produce a specific, preselected glycoform. In order to achieve glycan uniformity on the target protein, No Open Reading Frame (NORF) yeast cell lines are screened extensively during various stages of glycoengineering. In the absence of the target protein of interest, screening the NORF yeast cell lines for glycoform uniformity becomes a challenge. The common approach so far has been to analyze the total cell glycan pool released from glycoproteins of the NORF yeast cells to predict the N-glycan uniformity. As this does not always accurately predict the N-glycan end product, we describe in this chapter a detailed protocol for a non-affinity-based high-throughput purification of an endogenous glycoprotein. This protein of interest has been introduced during the early stages of glycoengineering process and its N-glycan profile is utilized as a tool for glycoengineering screening.


Subject(s)
Glycoproteins/isolation & purification , High-Throughput Screening Assays/methods , Molecular Biology/methods , Polysaccharides/chemistry , Anion Exchange Resins/chemistry , Chromatography, Ion Exchange , Glycosylation , Humans , Pichia , Polysaccharides/metabolism , Protein Engineering
10.
J Biotechnol ; 157(1): 198-206, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22100268

ABSTRACT

Pichia pastoris is a methylotropic yeast that has gained great importance as an organism for protein expression in recent years. Here, we report the expression of recombinant human erythropoietin (rhEPO) in glycoengineered P. pastoris. We show that glycosylation fidelity is maintained in fermentation volumes spanning six orders of magnitude and that the protein can be purified to high homogeneity. In order to increase the half-life of rhEPO, the purified protein was coupled to polyethylene glycol (PEG) and then compared to the currently marketed erythropoiesis stimulating agent, Aranesp(®) (darbepoetin). In in vitro cell proliferation assays the PEGylated protein was slightly, and the non-PEGylated protein was significantly more active than comparator. Pharmacodynamics as well as pharmacokinetic activity of PEGylated rhEPO in animals was comparable to that of Aranesp(®). Taken together, our results show that glycoengineered P. pastoris is a suitable production host for rhEPO, yielding an active biologic that is comparable to those produced in current mammalian host systems.


Subject(s)
Erythropoietin/biosynthesis , Pichia/metabolism , Protein Engineering/methods , Animals , Cell Proliferation/drug effects , Darbepoetin alfa , Erythropoietin/analogs & derivatives , Erythropoietin/blood , Erythropoietin/genetics , Erythropoietin/pharmacokinetics , Erythropoietin/pharmacology , Female , Glycosylation , Humans , Male , Mice , Pichia/genetics , Polyethylene Glycols , Polysaccharides/chemistry , Rats, Sprague-Dawley , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics
11.
Glycobiology ; 21(12): 1616-26, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21840970

ABSTRACT

The methylotrophic yeast, Pichia pastoris, is an important organism used for the production of therapeutic proteins. However, the presence of fungal-like glycans, such as those containing ß-mannose (Man) linkages, can elicit an immune response or bind to Man receptors, thus reducing their efficacy. Recent studies have confirmed that P. pastoris has four genes from the ß-mannosyl transferase (BMT) family and that Bmt2p is responsible for the majority of ß-Man linkages on glycans. While expressing recombinant human erythropoietin (rhEPO) in a developmental glycoengineered strain devoid of BMT2 gene expression, cross-reactivity was observed with an antibody raised against host cell antigens. Treatment of the rhEPO with protein N-glycosidase F eliminated cross-reactivity, indicating that the antigen was associated with the glycan. Thorough analysis of the glycan profile of rhEPO demonstrated the presence of low amounts of α-1,2-mannosidase resistant high-Man glycoforms. In an attempt to eliminate the α-mannosidase resistant glycoforms, we used a systemic approach to genetically knock-out the remaining members of the BMT family culminating in a quadruple bmt2,4,1,3 knock-out strain. Data presented here conclude that the additive elimination of Bmt2p, Bmt3p and Bmt1p activities are required for total abolition of ß-Man-associated glycans and their related antigenicity. Taken together, the elimination of ß-Man containing glycoforms represents an important step forward for the Pichia production platform as a suitable system for the production of therapeutic glycoproteins.


Subject(s)
Mannose/chemistry , Pichia/chemistry , Polysaccharides/chemistry , Polysaccharides/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Carbohydrate Conformation , Cross Reactions , Erythropoietin/chemistry , Erythropoietin/genetics , Erythropoietin/isolation & purification , Erythropoietin/metabolism , Humans , Mannose/metabolism , Mannosyltransferases/genetics , Mannosyltransferases/metabolism , Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase/metabolism , Pichia/enzymology , Pichia/genetics , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification
12.
Yeast ; 28(3): 237-52, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21360735

ABSTRACT

To humanize the glycosylation pathway in the yeast Pichia pastoris, we developed several combinatorial genetic libraries and used them to properly localize active eukaryotic mannosidases and sugar transferases. Here we report the details of the fusion of up to 66 N-terminal targeting sequences of fungal type II membrane proteins to 33 catalytic domains of heterologous glycosylation enzymes. We show that while it is difficult to predict which leader/catalytic domain will result in the desired activity, analysis of the fusion protein libraries allows for the selection of the leader/catalytic domain combinations that function properly. This combinatorial approach, together with a high-throughput screening protocol, has allowed us to humanize the yeast glycosylation pathway to secrete human glycoprotein with complex N-glycosylation.


Subject(s)
Endoplasmic Reticulum/enzymology , Glucosyltransferases/metabolism , Golgi Apparatus/enzymology , Mannosidases/metabolism , Pichia/enzymology , Protein Engineering , Glucosyltransferases/genetics , Mannosidases/genetics , Pichia/genetics , Protein Sorting Signals/genetics , Protein Transport , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
13.
Curr Opin Biotechnol ; 18(5): 387-92, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17951046

ABSTRACT

Yeasts have been extensively used as model organisms to elucidate cellular processes and their mechanism in lower eukaryotes. Consequently, a large number of powerful genetic tools have been developed to engineer yeast and improve its utility. These tools and the development of efficient large-scale fermentation processes have made recombinant protein expression in yeast an attractive choice. However, for the production of glycoproteins for human use, native high-mannose yeast glycosylation is not suitable and therefore represents a major limitation for yeast based protein expression systems. Over the last two decades several groups have attempted to overcome this problem, yet with limited success. Recently however, major advances in the glycoengineering of the yeast Pichia pastoris, have culminated in the production of fully humanized sialylated glycoproteins.


Subject(s)
Glycoproteins/metabolism , Pichia/physiology , Protein Engineering/trends , Recombinant Proteins/metabolism , Transfection/methods , Animals , Genetic Enhancement/methods , Glycoproteins/genetics , Glycosylation , Humans
14.
Science ; 313(5792): 1441-3, 2006 Sep 08.
Article in English | MEDLINE | ID: mdl-16960007

ABSTRACT

Yeast is a widely used recombinant protein expression system. We expanded its utility by engineering the yeast Pichia pastoris to secrete human glycoproteins with fully complex terminally sialylated N-glycans. After the knockout of four genes to eliminate yeast-specific glycosylation, we introduced 14 heterologous genes, allowing us to replicate the sequential steps of human glycosylation. The reported cell lines produce complex glycoproteins with greater than 90% terminal sialylation. Finally, to demonstrate the utility of these yeast strains, functional recombinant erythropoietin was produced.


Subject(s)
Erythropoietin/metabolism , Pichia/genetics , Protein Engineering , Sialoglycoproteins/biosynthesis , Animals , Cell Line , Cloning, Molecular , Cytidine Monophosphate N-Acetylneuraminic Acid/metabolism , Erythropoietin/chemistry , Erythropoietin/genetics , Genetic Vectors , Glycosylation , Humans , Pichia/metabolism , Rats , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Sialic Acids/metabolism , Sialoglycoproteins/chemistry , Sialoglycoproteins/genetics , Transformation, Genetic
15.
Nat Biotechnol ; 24(2): 210-5, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16429149

ABSTRACT

As the fastest growing class of therapeutic proteins, monoclonal antibodies (mAbs) represent a major potential drug class. Human antibodies are glycosylated in their native state and all clinically approved mAbs are produced by mammalian cell lines, which secrete mAbs with glycosylation structures that are similar, but not identical, to their human counterparts. Glycosylation of mAbs influences their interaction with immune effector cells that kill antibody-targeted cells. Here we demonstrate that human antibodies with specific human N-glycan structures can be produced in glycoengineered lines of the yeast Pichia pastoris and that antibody-mediated effector functions can be optimized by generating specific glycoforms. Glycoengineered P. pastoris provides a general platform for producing recombinant antibodies with human N-glycosylation.


Subject(s)
Antibodies, Monoclonal/biosynthesis , Genetic Enhancement/methods , Immunoglobulin G/biosynthesis , Immunoglobulin G/genetics , Pichia/genetics , Pichia/metabolism , Protein Engineering/methods , Antibodies, Monoclonal/genetics , Glycosylation , Humans , Recombinant Proteins/biosynthesis
16.
Glycobiology ; 15(6): 615-24, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15677381

ABSTRACT

Rat endomannosidase is a glycosidic enzyme that catalyzes the cleavage of di-, tri-, or tetrasaccharides (Glc(1-3)Man), from N-glycosylation intermediates with terminal glucose residues. To date it is the only characterized member of this class of endomannosidic enzymes. Although this protein has been demonstrated to localize to the Golgi lumenal membrane, the mechanism by which this occurs has not yet been determined. Using the rat endomannosidase sequence, we identified three homologs, one each in the human, mouse, and rat genomes. Alignment of the four encoded protein sequences demonstrated that the newly identified sequences are highly conserved but differed significantly at the N-terminus from the previously reported protein. In this study we have cloned two novel endomannosidase sequences from rat and human cDNA libraries, but were unable to amplify the open reading frame of the previously reported rat sequence. Analysis of the rat genome confirmed that the 59- and 39-termini of the previously reported sequence were in fact located on different chromosomes. This, in combination with our inability to amplify the previously reported sequence, indicated that the N-terminus of the rat endomannosidase sequence previously published was likely in error (a cloning artifact), and that the sequences reported in the current study encode the intact proteins. Furthermore, unlike the previous sequence, the three ORFs identified in this study encode proteins containing a single N-terminal transmembrane domain. Here we demonstrate that this region is responsible for Golgi localization and in doing so confirm that endomannosidase is a type II membrane protein, like the majority of other secretory pathway glycosylation enzymes.


Subject(s)
Mannosidases/genetics , Membrane Proteins/genetics , Amino Acid Sequence , Animals , Cloning, Molecular , Gene Expression Regulation, Enzymologic , Genetic Vectors/genetics , Humans , Mannosidases/biosynthesis , Mannosidases/classification , Membrane Proteins/biosynthesis , Membrane Proteins/classification , Mice , Molecular Sequence Data , Pichia/genetics , Rats , Sequence Alignment
17.
Glycobiology ; 14(9): 757-66, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15190003

ABSTRACT

A significant percentage of eukaryotic proteins contain posttranslational modifications, including glycosylation, which are required for biological function. However, the understanding of the structure-function relationships of N-glycans has lagged significantly due to the microheterogeneity of glycosylation in mammalian produced proteins. Recently we reported on the cellular engineering of yeast to replicate human N-glycosylation for the production of glycoproteins. Here we report the engineering of an artificial glycosylation pathway in Pichia pastoris blocked in dolichol oligosaccharide assembly. The PpALG3 gene encoding Dol-P-Man:Man(5)GlcNAc(2)-PP-Dol mannosyltransferase was deleted in a strain that was previously engineered to produce hybrid GlcNAcMan(5)GlcNAc(2) human N-glycans. Employing this approach, combined with the use of combinatorial genetic libraries, we engineered P. pastoris strains that synthesize complex GlcNAc(2)Man(3)GlcNAc(2) N-glycans with striking homogeneity. Furthermore, through expression of a Golgi-localized fusion protein comprising UDP-glucose 4-epimerase and beta-1,4-galactosyl transferase activities we demonstrate that this structure is a substrate for highly efficient in vivo galactose addition. Taken together, these data demonstrate that the artificial in vivo glycoengineering of yeast represents a major advance in the production of glycoproteins and will emerge as a practical tool to systematically elucidate the structure-function relationship of N-glycans.


Subject(s)
Galactose/metabolism , Glycoproteins/metabolism , Oligosaccharides/chemistry , Pichia/metabolism , Base Sequence , DNA Primers , Glycoproteins/chemistry , Glycoproteins/genetics , Glycosylation , Humans , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
18.
Glycobiology ; 14(5): 399-407, 2004 May.
Article in English | MEDLINE | ID: mdl-15033937

ABSTRACT

N-glycans are synthesized in both yeast and mammals through the ordered assembly of a lipid-linked core Glc(3)Man(9)GlcNAc(2) structure that is subsequently transferred to a nascent protein in the endoplasmic reticulum. Once folded, glycoproteins are then shuttled to the Golgi, where additional but divergent processing occurs in mammals and fungi. We cloned the Pichia pastoris homolog of the ALG3 gene, which encodes the enzyme that converts Man(5)GlcNAc(2)-Dol-PP to Man(6)GlcNAc(2)-Dol-PP. Deletion of this gene in an och1 mutant background resulted in the secretion of glycoproteins with a predicted Man(5)GlcNAc(2) structure that could be trimmed to Man(3)GlcNAc(2) by in vitro alpha-1,2-mannosidase treatment. However, several larger glycans ranging from Hex(6)GlcNAc(2) to Hex(12)GlcNAc(2) were also observed that were recalcitrant to an array of mannosidase digests. These results contrast the far simpler glycan profile found in Saccharomyces cerevisiae alg3-1 och1, indicating diverging Golgi processing in these two closely related yeasts. Finally, analysis of the P. pastoris alg3 deletion mutant in the presence and absence of the outer chain initiating Och1p alpha-1,6-mannosyltransferase activity suggests that the PpOch1p has a broader substrate specificity compared to its S. cerevisiae counterpart.


Subject(s)
Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , Mannosyltransferases/genetics , Membrane Proteins/genetics , Oligosaccharides/metabolism , Pichia/enzymology , Saccharomyces cerevisiae Proteins/genetics , Amino Acid Sequence , Carbohydrate Conformation , Mannose/metabolism , Mannosidases/metabolism , Mannosyltransferases/metabolism , Membrane Proteins/metabolism , Molecular Sequence Data , Pichia/genetics , Polysaccharides/metabolism , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/metabolism , Sequence Homology, Amino Acid
19.
Science ; 301(5637): 1244-6, 2003 Aug 29.
Article in English | MEDLINE | ID: mdl-12947202

ABSTRACT

We report the humanization of the glycosylation pathway in the yeast Pichia pastoris to secrete a human glycoprotein with uniform complex N-glycosylation. The process involved eliminating endogenous yeast glycosylation pathways, while properly localizing five active eukaryotic proteins, including mannosidases I and II, N-acetylglucosaminyl transferases I and II, and uridine 5'-diphosphate (UDP)-N-acetylglucosamine transporter. Targeted localization of the enzymes enabled the generation of a synthetic in vivo glycosylation pathway, which produced the complex human N-glycan N-acetylglucosamine2-mannose3-N-acetylglucosamine2 (GlcNAc2Man3GlcNAc2). The ability to generate human glycoproteins with homogeneous N-glycan structures in a fungal host is a step toward producing therapeutic glycoproteins and could become a tool for elucidating the structure-function relation of glycoproteins.


Subject(s)
Genetic Engineering , Glycoproteins/biosynthesis , Mannosidases/genetics , Pichia/genetics , Polysaccharides/metabolism , Recombinant Proteins/biosynthesis , Animals , Catalytic Domain , Endoplasmic Reticulum/metabolism , Glycoproteins/chemistry , Glycoproteins/genetics , Glycosylation , Golgi Apparatus/metabolism , Humans , Mannosidases/metabolism , Membrane Transport Proteins/metabolism , N-Acetylglucosaminyltransferases/metabolism , Peptide Library , Pichia/enzymology , Pichia/metabolism , Polysaccharides/chemistry , Protein Processing, Post-Translational , Protein Transport , Recombinant Fusion Proteins/metabolism , Transformation, Genetic
20.
Proc Natl Acad Sci U S A ; 100(9): 5022-7, 2003 04 29.
Article in English | MEDLINE | ID: mdl-12702754

ABSTRACT

The secretory pathway of Pichia pastoris was genetically re-engineered to perform sequential glycosylation reactions that mimic early processing of N-glycans in humans and other higher mammals. After eliminating nonhuman glycosylation by deleting the initiating alpha-1,6-mannosyltransferase gene from P. pastoris, several combinatorial genetic libraries were constructed to localize active alpha-1,2-mannosidase and human beta-1,2-N-acetylglucosaminyltransferase I (GnTI) in the secretory pathway. First, >32 N-terminal leader sequences of fungal type II membrane proteins were cloned to generate a leader library. Two additional libraries encoding catalytic domains of alpha-1,2-mannosidases and GnTI from mammals, insects, amphibians, worms, and fungi were cloned to generate catalytic domain libraries. In-frame fusions of the respective leader and catalytic domain libraries resulted in several hundred chimeric fusions of fungal targeting domains and catalytic domains. Although the majority of strains transformed with the mannosidase/leader library displayed only modest in vivo [i.e., low levels of mannose (Man)(5)-(GlcNAc)(2)] activity, we were able to isolate several yeast strains that produce almost homogeneous N-glycans of the (Man)(5)-(GlcNAc)(2) type. Transformation of these strains with a UDP-GlcNAc transporter and screening of a GnTI leader fusion library allowed for the isolation of strains that produce GlcNAc-(Man)(5)-(GlcNAc)(2) in high yield. Recombinant expression of a human reporter protein in these engineered strains led to the formation of a glycoprotein with GlcNAc-(Man)(5)-(GlcNAc)(2) as the primary N-glycan. Here we report a yeast able to synthesize hybrid glycans in high yield and open the door for engineering yeast to perform complex human-like glycosylation.


Subject(s)
Pichia/genetics , Protein Engineering , Recombinant Fusion Proteins/genetics , Base Sequence , DNA Primers , Endoplasmic Reticulum/enzymology , Endoplasmic Reticulum/metabolism , Glycosylation , Golgi Apparatus/enzymology , Golgi Apparatus/metabolism , Humans , Mannosyltransferases/genetics , Pichia/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
SELECTION OF CITATIONS
SEARCH DETAIL