Publications

 

2022

The Golgi complex (GC) is a fundamental element of the secretory pathway (Glick and Nakano, 2009). In vertebrate cells, the GC is generally organized in the form of functionally polarized stacks of cisternae, each containing a distinct set of cargo-processing enzymes (Wei and Seemann, 2010). An additional level of organization is present in mammalian cells, where adjacent stacks are connected by membranous tubules (Rambourg and Clermont, 1990) to form a single copy organelle (Cole et al., 1996), called Golgi ribbon, often in close proximity to the centrosome (Wei and Seemann, 2010). The proteins and lipids that are synthesized in the endoplasmic reticulum (ER) are transported to the cis-Golgi through an intermediate compartment (Appenzeller-Herzog and Hauri, 2006). The cargoes are then processed through the GC and sorted at the trans-Golgi network (TGN) for transport to specific plasma membrane domains or organelles (Glick and Nakano, 2009). As it is oft-repeated, the Golgi apparatus (and secretory pathway) is responsible for producing and positioning nearly one-third of the proteome and thus is a primary organizer of the mammalian cell. This is a process in which Golgi-resident glycosyltransferases play a major role by generating protein diversity and affecting many physiological and pathological processes (Pinho and Reis, 2015; Bellis et al., 2022). 

Modeling glycan biosynthesis is becoming increasingly important due to the far-reaching implications that glycosylation can exhibit, from pathologies to biopharmaceutical manufacturing. Here we describe a stochastic simulation approach, to overcome the deterministic nature of previous models, that aims to simulate the action of glycan modifying enzymes to produce a glycan profile. This is then coupled with an approximate Bayesian computation methodology to systematically fit to empirical data in order to determine which set of parameters adequately describes the organization of enzymes within the Golgi. The model is described in detail along with a proof of concept and therapeutic applications.

2020

Nanoparticles could conceal bioactive proteins during therapeutic delivery, avoiding side effects. Superparamagnetic iron oxide nanoparticles (SPIONs) coated with a temperature-sensitive polymer were tested for protein release. We show that coated SPIONs can entrap test proteins and release them in a temperature-controlled manner in a biological system. Magnetically heating SPIONs triggered protein release at bulk solution temperatures below the polymer transition. The entrapped growth factor Wnt3a was inactive until magnetically triggered release, upon which it could increase mesenchymal stem cell proliferation. Once the polymer transition will be chemically adjusted above body temperature, this system could be used for targeted cell stimulation in model animals and humans. 

2019

Heterogeneity is an inherent feature of the glycosylation process. Mammalian cells often produce a variety of glycan structures on separate molecules of the same protein, known as glycoforms. This heterogeneity is not random but is controlled by the organization of the glycosylation machinery in the Golgi cisternae. In this work, we use a computational model of the N-glycosylation process to probe how the organization of the glycosylation machinery into different cisternae drives N-glycan biosynthesis toward differing degrees of heterogeneity. Using this model, we demonstrate the N-glycosylation potential and limits of the mammalian Golgi apparatus, for example how the number of cisternae limits the goal of achieving near homogeneity for N-glycans. The production of specific glycoforms guided by this computational study could pave the way for “glycoform engineering,” which will find uses in the functional investigation of glycans, the modulation of glycan-mediated physiological functions, and in biotechnology.

The decoration of proteins by carbohydrates is essential for eukaryotic life yet heterogeneous due to a lack of biosynthetic templates. This complex carbohydrate mixture—the glycan profile—is generated in the compartmentalized Golgi, in which level and localization of glycosylation enzymes are key determinants. Here, we develop and validate a computational model for glycan biosynthesis to probe how the biosynthetic machinery creates different glycan profiles. We combined stochastic modeling with Bayesian fitting that enables rigorous comparison to experimental data despite starting with uncertain initial parameters. This is an important development in the field of glycan modeling, which revealed biological insights about the glycosylation machinery in altered cellular states. We experimentally validated changes in N-linked glycan-modifying enzymes in cells with perturbed intra-Golgi-enzyme sorting and the predicted glycan-branching activity during osteogenesis. Our model can provide detailed information on altered biosynthetic paths, with potential for advancing treatments for glycosylation-related diseases and glyco-engineering of cells. 

COPI is a key mediator of protein trafficking within the secretory pathway. COPI is recruited to the membrane primarily through binding to Arf GTPases, upon which it undergoes assembly to form coated transport intermediates responsible for trafficking numerous proteins, including Golgi-resident enzymes. Here, we identify GORAB, the protein mutated in the skin and bone disorder gerodermia osteodysplastica, as a component of the COPI machinery. GORAB forms stable domains at the trans-Golgi that, via interactions with the COPI-binding protein Scyl1, promote COPI recruitment to these domains. Pathogenic GORAB mutations perturb Scyl1 binding or GORAB assembly into domains, indicating the importance of these interactions. Loss of GORAB causes impairment of COPI-mediated retrieval of trans-Golgi enzymes, resulting in a deficit in glycosylation of secretory cargo proteins. Our results therefore identify GORAB as a COPI scaffolding factor, and support the view that defective protein glycosylation is a major disease mechanism in gerodermia osteodysplastica.

2018

Glycans are inherently heterogeneous, yet glycosylation is essential in eukaryotes, and glycans show characteristic cell type-dependent distributions. By using an immortalized human mesenchymal stromal cell (MSC) line model, we show that both N- and O-glycan processing in the Golgi functionally modulates early steps of osteogenic differentiation. We found that inhibiting O-glycan processing in the Golgi prior to the start of osteogenesis inhibited the mineralization capacity of the formed osteoblasts 3 weeks later. In contrast, inhibition of N-glycan processing in MSCs altered differentiation to enhance the mineralization capacity of the osteoblasts. The effect of N-glycans on MSC differentiation was mediated by the phosphoinositide-3-kinase (PI3K)/Akt pathway owing to reduced Akt phosphorylation. Interestingly, by inhibiting PI3K during the first 2 days of osteogenesis, we were able to phenocopy the effect of inhibiting N-glycan processing. Thus, glycan processing provides another layer of regulation that can modulate the functional outcome of differentiation. Glycan processing can thereby offer a novel set of targets for many therapeutically attractive processes.

2017

A method has been developed for release/isolation of O-glycans from glycoproteins in whole cell lysates for mass spectrometric analysis. Cells are lysed in SDS, which is then exchanged for urea and ammonium bicarbonate in a centrifugal filter, before treating with NH4OH to release O-glycans. Following centrifugation, O-glycans are recovered in the filtrate. Sonication achieves O-glycan release in 1 h. Combining the established protocol for filter-aided N-glycan separation, here optimized for enhanced PNGase F efficiency, with the developed O-glycan release method allows analysis of both N- and O-glycans from one sample, in the same filter unit, from 0.5 to 1 million cells. The method is compatible with subsequent analysis of the residual protein by liquid chromatography–mass spectrometry (LC–MS) after glycan release. The medium throughput approach is amenable to analysis of biological replicates, offering a simple way to assess the often subtle changes to glycan profiles accompanying differentiation and disease progression, in a statistically robust way.

Tumor cells gain metastatic capacity through a Golgi phosphoprotein 3–dependent (GOLPH3-dependent) Golgi membrane dispersal process that drives the budding and transport of secretory vesicles. Whether Golgi dispersal underlies the pro-metastatic vesicular trafficking that is associated with epithelial-to-mesenchymal transition (EMT) remains unclear. Here, we have shown that, rather than causing Golgi dispersal, EMT led to the formation of compact Golgi organelles with improved ribbon linking and cisternal stacking. Ectopic expression of the EMT-activating transcription factor ZEB1 stimulated Golgi compaction and relieved microRNA-mediated repression of the Golgi scaffolding protein PAQR11. Depletion of PAQR11 dispersed Golgi organelles and impaired anterograde vesicle transport to the plasma membrane as well as retrograde vesicle tethering to the Golgi. The N-terminal scaffolding domain of PAQR11 was associated with key regulators of Golgi compaction and vesicle transport in pull-down assays and was required to reconstitute Golgi compaction in PAQR11-deficient tumor cells. Finally, high PAQR11 levels were correlated with EMT and shorter survival in human cancers, and PAQR11 was found to be essential for tumor cell migration and metastasis in EMT-driven lung adenocarcinoma models. We conclude that EMT initiates a PAQR11-mediated Golgi compaction process that drives metastasis.

2000 - 2016

2016

Fari, K, S Takacs, D Ungar, R Sinka (2016) The role of acroblast formation during Drosophila spermatogenesis. Biol. Open, 5, 1102-1110

Ha, JY, HT Chou, D Ungar, CK Yip, T Walz, FM Hughson (2016) Molecular architecture of the complete COG tethering complex. Nat. Struct. Mol. Biol., 23, 758-760, doi:10.1038/nsmb.3263

Wilson, KM, JE Thomas-Oates, P Genever, D Ungar (2016) Glycan profiling shows unvaried N-glycomes in MSC clones with distinct differentiation potentials. Front. Cell Dev. Biol., 4:52, doi: 10.3389/fcell.2016.00052

Bailey Blackburn J, I Pokrovskaya, P Fisher, D Ungar, V Lupashin (2016) COG Complex complexities: Detailed characterization of a complete set of HEK293T cells lacking individual COG subunits. Front. Cell Dev. Biol.,4:23, doi.org/10.3389/fcell.2016.00023

Fisher P, D Ungar (2016) Bridging the gap between glycosylation and vesicle traffic. Front. Cell Dev. Biol., 4:15, doi: 10.3389/fcell.2016.0001

2014

Abdul Rahman S, E Bergström, CJ Watson, KM Wilson, DA Ashford, JR Thomas, D Ungar*, JE Thomas-Oates (2014) Filter-Aided N-Glycan Separation (FANGS): a Convenient Sample Preparation Method for Mass Spectrometric N-Glycan Profiling. J. Prot. Res., 13, 1167–1176 

Cottam NP, KM Wilson, BG Ng, C Körner, HH Freeze, D Ungar (2014) Dissecting functions of the conserved oligomeric Golgi tethering complex using a cell free assay. Traffic, 15, 12-21 

2013

Willett R, D Ungar*, V Lupashin (2013) The Golgi puppet master: COG complex at center stage of membrane trafficking interactions. Histochem. Cell Biol., 140 (3), 271-283, doi:10.1007/s00418-013-1117-6 

Willett R, Kudlyk T, Pokrovskaya I, Schönherr R, Ungar D, Duden R, Lupalshin V (2013) COG complexes form spatial landmarks for distinct SNARE complexes. Nat. Commun., 4: 1553 doi:10.1038/ncomms2535 

Miller VJ, P Sharma, TA Kudlyk, L Frost, AP Rofe, IJ Watson, R Duden, M Lowe, VV Lupashin, D Ungar (2013) Molecular insights into vesicle tethering at the Golgi by the Conserved Oligomeric Golgi (COG) complex and the golgin TMF. J. Biol. Chem., 288, 4229-4240 

2012

Miller, VJ, D Ungar (2012) Re’COG’nition at the Golgi. Traffic, 13, 891-897

Cottam, NP, D Ungar (2012) Retrograde vesicle transport in the Golgi. Protoplasma, 249(4), 943-955 

2010

Lübbehusen, J, C Thiel, N Rind, D Ungar, BH Prinsen, TJ de Koning, PM van Hasselt and C Körner (2010) Fatal outcome due to deficiency of subunit 6 of the conserved oligomeric Golgi complex leading to a new type of congenital disorders of glycosylation. Hum. Mol. Genet., 19, 3623-33 

2009

Ungar, D (2009) Golgi linked protein glycosylation and associated diseases, Sem. Cell Dev. Biol., 20, 762-769 

Richardson, BC, R Smith, D Ungar, A Nakamura, P Jeffrey, VV Lupashin and FM Hughson (2009) Structural basis for a human glycosylation disorder caused by mutation of the COG4 gene, PNAS, 106, 13329-34 

2008

Lupashin, VV and D Ungar (2008) COG complex. In: The Golgi Apparatus (State of the art 110 years after Camillo Golgi's discovery), eds. Mironov and Pavelka, Springer Verlag, Wien, chapter 2.7 

2007

Foulquier, F, D Ungar, E Reynders, R Zeevaert, P Mills, MT García-Silva, P Briones, B Winchester, W Morelle, M Krieger, W Annaert and G Matthijs (2007) A new inborn error of glycosylation due to a Cog8 deficiency reveals a critical role for the Cog1-Cog8 interaction in COG complex formation. Hum. Mol. Genet., 16, 717-730

Kranz, C, BG Ng, L Sun, V Sharma, EA Eklund, Y Miura, D Ungar, V Lupashin, DR Winkel, JF Cipollo, CE Costello, E Loh, W Hong and HH Freeze (2007) COG8 deficiency causes new Congenital Disorder of Glycosylation type IIh. Hum. Mol. Genet., 16, 731-741 

Ng, BG, C Kranz,  EEO Hagebeuk, M Duran, NGGM Abeling, B Wuyts, D Ungar, V Lupashin,  CM Hartdorff, BT Poll-The, HH Freeze (2007) Molecular and Clinical Characterization of a Moroccan Cog7 Deficient Patient. Mol. Genet. Metab., 91, 201-204 

Cavanaugh, L, X Chen, BC Richardson, D Ungar, I Pelczer, J Rizo and FM Hughson (2007) The structural core of Cog2p reveals architectural similarity to exocyst subunits. J. Biol. Chem., 282, 23418-23426 

2006

Ungar, D, T Oka, M Krieger and FM Hughson (2006) Retrograde transport on the COG railway. Trends Cell Biol., 16, 113-120

2005   

Ungar, D, T Oka, E Vasile, M Krieger and FM Hughson (2005) Subunit architecture of the conserved oligomeric Golgi complex. J. Biol. Chem., 280, 32729-32735 

Oka, T, E Vasile, M Penman, CD Novina, DM Dykxhoorn, D Ungar, FM Hughson and M Krieger (2005) Genetic analysis of the subunit organization and function of the COG complex: Studies of COG5 and COG7 deficient mammalian cells. J. Biol. Chem., 280, 32736-32745 

2004   

Oka, T, D Ungar, FM Hughson and M Krieger (2004) The COG and COPI complexes interact to control the abundance of GEARs, a subset of Golgi integral membrane proteins. Mol. Biol. Cell, 15, 2423-2435 

2003

Ungar, D and FM Hughson (2003) SNARE Protein Structure and Function. Annu. Rev. Cell Dev. Biol., 19, 493-517

2002

Ungar, D, T Oka, EE Brittle, E Vasile, VV Lupashin, JE Chatterton, JE Heuser, M Krieger and MG Waters (2002) Characterization of a mammalian Golgi-localized protein complex, COG, that is required for normal Golgi morphology and function. J. Cell Biol., 157, 405-415

2001   

Ungar, D, A Barth, W Haase, A Kaunzinger, E Lewitzki, T Ruiz, H Reiländer and H Michel (2001) Analysis of a putative voltage-gated prokaryotic potassium channel. Eur. J. Biochem., 268, 5386-5396