Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Proc Natl Acad Sci U S A ; 117(12): 6675-6685, 2020 03 24.
Article in English | MEDLINE | ID: mdl-32152119

ABSTRACT

A comprehensive understanding of the development and evolution of human B cell responses induced by pathogen exposure will facilitate the design of next-generation vaccines. Here, we utilized a high-throughput single B cell cloning technology to longitudinally track the human B cell response to the yellow fever virus 17D (YFV-17D) vaccine. The early memory B cell (MBC) response was mediated by both classical immunoglobulin M (IgM) (IgM+CD27+) and switched immunoglobulin (swIg+) MBC populations; however, classical IgM MBCs waned rapidly, whereas swIg+ and atypical IgM+ and IgD+ MBCs were stable over time. Affinity maturation continued for 6 to 9 mo following vaccination, providing evidence for the persistence of germinal center activity long after the period of active viral replication in peripheral blood. Finally, a substantial fraction of the neutralizing antibody response was mediated by public clones that recognize a fusion loop-proximal antigenic site within domain II of the viral envelope glycoprotein. Overall, our findings provide a framework for understanding the dynamics and complexity of human B cell responses elicited by infection and vaccination.


Subject(s)
Antibodies, Viral/immunology , Antigens, Viral/immunology , B-Lymphocytes/immunology , Immunologic Memory/immunology , Yellow Fever Vaccine/immunology , Yellow Fever/prevention & control , Yellow fever virus/immunology , Adult , Humans , Vaccination , Vaccines, Attenuated/immunology , Viral Envelope Proteins/immunology , Virus Replication , Yellow Fever/immunology , Yellow Fever/virology , Yellow Fever Vaccine/administration & dosage
2.
Proc Natl Acad Sci U S A ; 114(5): 944-949, 2017 01 31.
Article in English | MEDLINE | ID: mdl-28096333

ABSTRACT

Antibodies are a highly successful class of biological drugs, with over 50 such molecules approved for therapeutic use and hundreds more currently in clinical development. Improvements in technology for the discovery and optimization of high-potency antibodies have greatly increased the chances for finding binding molecules with desired biological properties; however, achieving drug-like properties at the same time is an additional requirement that is receiving increased attention. In this work, we attempt to quantify the historical limits of acceptability for multiple biophysical metrics of "developability." Amino acid sequences from 137 antibodies in advanced clinical stages, including 48 approved for therapeutic use, were collected and used to construct isotype-matched IgG1 antibodies, which were then expressed in mammalian cells. The resulting material for each source antibody was evaluated in a dozen biophysical property assays. The distributions of the observed metrics are used to empirically define boundaries of drug-like behavior that can represent practical guidelines for future antibody drug candidates.


Subject(s)
Antibodies, Monoclonal , Drug Discovery/methods , Amino Acid Sequence , Antibodies, Monoclonal/chemistry , Biophysical Phenomena , Drug Approval , HEK293 Cells , Humans , Immunoglobulin G/chemistry
3.
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
4.
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
5.
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
6.
J Ind Microbiol Biotechnol ; 37(9): 961-71, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20711797

ABSTRACT

The methylotrophic yeast Pichia pastoris has recently been engineered to express therapeutic glycoproteins with uniform human N-glycans at high titers. In contrast to the current art where producing therapeutic proteins in mammalian cell lines yields a final product with heterogeneous N-glycans, proteins expressed in glycoengineered P. pastoris can be designed to carry a specific, preselected glycoform. However, significant variability exists in fermentation performance between genotypically similar clones with respect to cell fitness, secreted protein titer, and glycan homogeneity. Here, we describe a novel, multidimensional screening process that combines high and medium throughput tools to identify cell lines producing monoclonal antibodies (mAbs). These cell lines must satisfy multiple selection criteria (high titer, uniform N-glycans and cell robustness) and be compatible with our large-scale production platform process. Using this selection process, we were able to isolate a mAb-expressing strain yielding a titer (after protein A purification) in excess of 1 g/l in 0.5-l bioreactors.


Subject(s)
Antibodies, Monoclonal/biosynthesis , Genetic Engineering , Glycoproteins/biosynthesis , Pichia/isolation & purification , Recombinant Proteins/biosynthesis , Antibodies, Monoclonal/genetics , Bioreactors , Cell Culture Techniques , Cell Line , DNA, Fungal/genetics , Fermentation , Gene Expression , Glycoproteins/genetics , Glycosylation , Humans , Microbiological Techniques , Pichia/genetics , Pichia/metabolism , Recombinant Proteins/genetics , Selection, Genetic , Transformation, Genetic
7.
PLoS One ; 15(3): e0229206, 2020.
Article in English | MEDLINE | ID: mdl-32134960

ABSTRACT

Here we describe an industry-wide collaboration aimed at assessing the binding properties of a comprehensive panel of monoclonal antibodies (mAbs) against programmed cell death protein 1 (PD-1), an important checkpoint protein in cancer immunotherapy and validated therapeutic target, with well over thirty unique mAbs either in clinical development or market-approved in the United States, the European Union or China. The binding kinetics of the PD-1/mAb interactions were measured by surface plasmon resonance (SPR) using a Carterra LSA instrument and the results were compared to data collected on a Biacore 8K. The effect of chip type on the SPR-derived binding rate constants and affinities were explored and the results compared with solution affinities from Meso Scale Discovery (MSD) and Kinetic Exclusion Assay (KinExA) experiments. When using flat chip types, the LSA and 8K platforms yielded near-identical kinetic rate and affinity constants that matched solution phase values more closely than those produced on 3D-hydrogels. Of the anti-PD-1 mAbs tested, which included a portion of those known to be in clinical development or approved, the affinities spanned from single digit picomolar to nearly 425 nM, challenging the dynamic range of our methods. The LSA instrument was also used to perform epitope binning and ligand competition studies which revealed over ten unique competitive binding profiles within this group of mAbs.


Subject(s)
Antibodies, Monoclonal/pharmacology , Biosensing Techniques/methods , Programmed Cell Death 1 Receptor/immunology , China , Drug Development , Epitopes/immunology , European Union , High-Throughput Screening Assays , Humans , Programmed Cell Death 1 Receptor/chemistry , Protein Binding , Surface Plasmon Resonance , United States
8.
bioRxiv ; 2020 May 16.
Article in English | MEDLINE | ID: mdl-32511337

ABSTRACT

Broadly protective vaccines against known and pre-emergent coronaviruses are urgently needed. Critical to their development is a deeper understanding of cross-neutralizing antibody responses induced by natural human coronavirus (HCoV) infections. Here, we mined the memory B cell repertoire of a convalescent SARS donor and identified 200 SARS-CoV-2 binding antibodies that target multiple conserved sites on the spike (S) protein. A large proportion of the antibodies display high levels of somatic hypermutation and cross-react with circulating HCoVs, suggesting recall of pre-existing memory B cells (MBCs) elicited by prior HCoV infections. Several antibodies potently cross-neutralize SARS-CoV, SARS-CoV-2, and the bat SARS-like virus WIV1 by blocking receptor attachment and inducing S1 shedding. These antibodies represent promising candidates for therapeutic intervention and reveal a new target for the rational design of pan-sarbecovirus vaccines.

9.
Science ; 369(6504): 731-736, 2020 08 07.
Article in English | MEDLINE | ID: mdl-32540900

ABSTRACT

Broadly protective vaccines against known and preemergent human coronaviruses (HCoVs) are urgently needed. To gain a deeper understanding of cross-neutralizing antibody responses, we mined the memory B cell repertoire of a convalescent severe acute respiratory syndrome (SARS) donor and identified 200 SARS coronavirus 2 (SARS-CoV-2) binding antibodies that target multiple conserved sites on the spike (S) protein. A large proportion of the non-neutralizing antibodies display high levels of somatic hypermutation and cross-react with circulating HCoVs, suggesting recall of preexisting memory B cells elicited by prior HCoV infections. Several antibodies potently cross-neutralize SARS-CoV, SARS-CoV-2, and the bat SARS-like virus WIV1 by blocking receptor attachment and inducing S1 shedding. These antibodies represent promising candidates for therapeutic intervention and reveal a target for the rational design of pan-sarbecovirus vaccines.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Betacoronavirus/immunology , Broadly Neutralizing Antibodies/immunology , Severe acute respiratory syndrome-related coronavirus/immunology , Spike Glycoprotein, Coronavirus/immunology , Adult , Aged , Angiotensin-Converting Enzyme 2 , Antibody Affinity , B-Lymphocyte Subsets/immunology , Binding Sites , Cross Reactions , Epitopes , Female , Humans , Immunologic Memory , Male , Middle Aged , Neutralization Tests , Peptidyl-Dipeptidase A/chemistry , Peptidyl-Dipeptidase A/metabolism , Protein Domains , Receptors, Coronavirus , Receptors, Virus/chemistry , Receptors, Virus/metabolism , SARS-CoV-2 , Severe Acute Respiratory Syndrome/immunology , Somatic Hypermutation, Immunoglobulin , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/metabolism , Young Adult
10.
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
11.
Appl Environ Microbiol ; 74(4): 1076-86, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18083888

ABSTRACT

The production by filamentous fungi of therapeutic glycoproteins intended for use in mammals is held back by the inherent difference in protein N-glycosylation and by the inability of the fungal cell to modify proteins with mammalian glycosylation structures. Here, we report protein N-glycan engineering in two Aspergillus species. We functionally expressed in the fungal hosts heterologous chimeric fusion proteins containing different localization peptides and catalytic domains. This strategy allowed the isolation of a strain with a functional alpha-1,2-mannosidase producing increased amounts of N-glycans of the Man5GlcNAc2 type. This strain was further engineered by the introduction of a functional GlcNAc transferase I construct yielding GlcNAcMan5GlcNac2 N-glycans. Additionally, we deleted algC genes coding for an enzyme involved in an early step of the fungal glycosylation pathway yielding Man3GlcNAc2 N-glycans. This modification of fungal glycosylation is a step toward the ability to produce humanized complex N-glycans on therapeutic proteins in filamentous fungi.


Subject(s)
Aspergillus/metabolism , Polysaccharides/biosynthesis , Protein Engineering/methods , Transformation, Bacterial/genetics , Base Sequence , Cloning, Molecular , DNA Primers/genetics , Gene Transfer Techniques , Mannosyltransferases/genetics , Molecular Sequence Data , Polysaccharides/genetics , Recombinant Fusion Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , alpha-Mannosidase/metabolism
12.
MAbs ; 9(2): 257-268, 2017.
Article in English | MEDLINE | ID: mdl-27937066

ABSTRACT

Monovalent bispecific antibodies (BsAbs) are projected to have broad clinical applications due to their ability to bind two different targets simultaneously. Although they can be produced using recombinant technologies, the correct pairing of heavy and light chains is a significant manufacturing problem. Various approaches exploit mutations or linkers to favor the formation of the desired BsAb, but a format using a single common light chain has the advantage that no other modification to the antibody is required. This strategy reduces the number of formed molecules to three (the BsAb and the two parent mAbs), but the separation of the BsAb from the two monovalent parent molecules still poses a potentially difficult purification challenge. Current methods employ ion exchange chromatography and linear salt gradients, but are only successful if the difference in the observed isoelectric points (pIs) of two parent molecules is relatively large. Here, we describe the use of highly linear pH gradients for the facile purification of common light chain BsAbs. The method is effective at separating molecules with differences in pI as little as 0.10, and differing in their sequence by only a single charged amino acid. We also demonstrate that purification resins validated for manufacturing are compatible with this approach.


Subject(s)
Antibodies, Bispecific/isolation & purification , Chromatography, Ion Exchange/methods , Immunoglobulin G/isolation & purification , Proton-Motive Force , Humans , Protein Engineering/methods
13.
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
14.
Bioeng Bugs ; 3(1): 32-7, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-22126802

ABSTRACT

The methylotropic yeast Pichia pastoris has been used for more than two decades to successfully produce a large number of recombinant proteins. Currently, a wide variety of auxotrophic and drug based selection markers are employed to screen for clones expressing the protein of interest. For most proteins an increased copy number of the integrated plasmid results in higher levels of expression, but these multi-copy integrants can be unstable due to the propensity of P. pastoris for homologous recombination. Here we describe a multi-copy selection system based on ade1 and ade2 auxotrophic parent strains and the respective attenuated markers with truncated promoter regions. We show that for all four proteins we tested, the use of the attenuated markers leads to increased protein expression when compared with selection based on the full strength markers. The fact that the adenine auxotrophic strains grow more slowly than the complemented counterparts essentially ensures the stability of multi-copy integration. At the same time, the accumulation of a red dye in the auxotrophic strains also provides an easy, color-based selection for transformants with multiple copies.


Subject(s)
Fungal Proteins/genetics , Pichia/genetics , Promoter Regions, Genetic/genetics , Plasmids/genetics
15.
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
16.
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
17.
Yeast ; 22(4): 295-304, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15789348

ABSTRACT

Screening of a partial genomic database of Pichia pastoris allowed us to identify the ARG1, ARG2, ARG3, HIS1, HIS2, HIS5 and HIS6 genes, based on homology to their Saccharomyces cerevisiae counterparts. Based on the cloned sequences, a set of disruption vectors was constructed, using the previously described PpURA5-blaster as a selectable marker, and the cloned genes were individually disrupted. All disruptants exhibited the expected auxotrophic phenotypes, with only the his2 knockouts displaying a bradytroph phenotype. To allow their use as auxotrophic markers, we amplified the open reading frames and respective promoters and terminator regions of PpARG1, PpARG2, PpARG3, PpHIS1, PpHIS2 and PpHIS5. We then designed a set of integration vectors harbouring cassettes of the ARG pathway as selectable markers, to disrupt the genes of the HIS pathway and vice versa. Employing this strategy, we devised a scheme allowing for the rapid and stable introduction of several heterologous genes into the genome of P. pastoris without the need for recyclable markers or strains with multiple auxotrophies. Furthermore, simple replica-plating, instead of cost-consuming and labour-intensive colony PCR or Southern analysis, can be used to identify positive transformants, making this approach amendable for initial high-throughput applications, which can then be followed up by a more careful analysis of the selected transformants.


Subject(s)
Arginine/biosynthesis , Genes, Fungal , Histidine/biosynthesis , Pichia/genetics , Arginine/genetics , Base Sequence , Biomarkers , Cloning, Molecular , DNA, Fungal/chemistry , DNA, Fungal/genetics , Histidine/genetics , Molecular Sequence Data , Mutagenesis, Insertional , Pichia/growth & development , Pichia/metabolism , Plasmids , Polymerase Chain Reaction , Sequence Analysis, DNA
18.
Yeast ; 20(15): 1279-90, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14618566

ABSTRACT

A pair of degenerate primers was used for amplification and cloning of a DNA fragment containing parts of the P. pastoris URA5 and SEC65 genes. Using additional information from a partial genomic sequence of P. pastoris, we cloned and sequenced a 1.9 kb chromosomal fragment containing the complete orotate-phosphoribosyltransferase-encoding URA5 gene. A disruption cassette was constructed by replacing a small part of the open reading frame with a kanamycin-resistance gene. The P. pastoris wild-type strain NRRL Y-11430 was transformed with the disruption cassette and an ura5 auxotrophic strain was identified. To generate marker constructs that can be reused in successive transformations of a single strain, we constructed two lacZ-PpURA3-lacZ and lacZ-PpURA5-lacZ cassettes and used them to disrupt PpOCH1. The PpURA3 and PpURA5 genes in the disruptants were then successfully recycled by selecting for resistance to 5'-fluoro-orotic acid. We also assembled a set of modular plasmids that can be used for the stable genetic modification of P. pastoris via a double cross-over event. The sequence presented here has been submitted to the EMBL data library under Accession No. AY303544.


Subject(s)
Orotate Phosphoribosyltransferase/genetics , Orotic Acid/analogs & derivatives , Pichia/enzymology , Pichia/genetics , Amino Acid Sequence , Base Sequence , Cloning, Molecular , DNA, Fungal/chemistry , DNA, Fungal/genetics , Drug Resistance, Fungal/genetics , Fungal Proteins/genetics , Genetic Vectors , Molecular Sequence Data , Mutagenesis, Insertional , Orotic Acid/pharmacology , Plasmids , Polymerase Chain Reaction , Sequence Alignment , Sequence Analysis, DNA , Transformation, Genetic/genetics
19.
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
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