key: cord-0683787-k7s2hu2h authors: D'Souza, Zinia; Taher, Farhana S.; Lupashin, Vladimir V. title: Golgi inCOGnito: From vesicle tethering to human disease date: 2020-07-27 journal: Biochim Biophys Acta Gen Subj DOI: 10.1016/j.bbagen.2020.129694 sha: 4dc09789321a102a56bafa7306512426cdf35006 doc_id: 683787 cord_uid: k7s2hu2h The Conserved Oligomeric Golgi (COG) complex, a multi-subunit vesicle tethering complex of the CATCHR (Complexes Associated with Tethering Containing Helical Rods) family, controls several aspects of cellular homeostasis by orchestrating retrograde vesicle traffic within the Golgi. The COG complex interacts with all key players regulating intra-Golgi trafficking, namely SNAREs, SNARE-interacting proteins, Rabs, coiled-coil tethers, and vesicular coats. In cells, COG deficiencies result in the accumulation of non-tethered COG-complex dependent (CCD) vesicles, dramatic morphological and functional abnormalities of the Golgi and endosomes, severe defects in N- and O- glycosylation, Golgi retrograde trafficking, sorting and protein secretion. In humans, COG mutations lead to severe multi-systemic diseases known as COG-Congenital Disorders of Glycosylation (COG-CDG). In this report, we review the current knowledge of the COG complex and analyze COG-related trafficking and glycosylation defects in COG-CDG patients. glycosylation machinery is currently unknown and likely requires several novel adaptor proteins. Apart from cytoplasmic sorting signals, length of the transmembrane domains (TMDs) of glycosylation enzymes might contribute to their retention and sorting within the Golgi. cis-Golgi resident proteins have shorter TMDs compared to late Golgi residents. Membrane thickness increases from ER to the PM due to the increasing concentration of sphingolipids and sterols. It is energetically favorable for shorter TMDs to be retained in thinner membranes and sorted into phospholipid-rich COPI vesicles rather than cholesterol-rich clathrin-coated vesicles targeted to the PM [16, 17] . Another proposed mechanism of Golgi enzyme retention is the formation of multimeric complexes. Sequentially acting glycosylation enzymes in the same cisternae can form dimers and higher-order oligomers. This increases the size of the protein preventing it from entering vesicles, leaving the Golgi. Oligomerization of sequentially acting enzymes also enhances the efficiency of glycosylation [18, 19] . Also, the luminal environment within the Golgi governs glycosylation and Golgi homeostasis. Each Golgi cisternae has a unique pH and ionic environment, which contributes to correct enzyme localization [20] and also regulates enzyme activity [21] . The contribution of these factors to Golgi homeostasis has been reviewed in Kellokumpu [22] . Furthermore, reduced luminal volume in flattened Golgi stacks facilitates enzymes-substrate encounters [23] . Multi-subunit tethering complexes (MTCs) are a group of evolutionarily conserved vesicular tethers consisting of three to ten subunits. They are loosely sub-grouped into CATCHRs and non-CATCHRs [24] . CATCHR family includes MTCs functioning along the secretory pathway, namely, Dsl1/ZW10 complex, COG, GARP, EARP and the exocyst; non-CATCHR group includes CORVET and HOPS complexes which are involved in the endosomal trafficking. TRAPP, a unique MTC in structure and function is involved in ER to Golgi trafficking and autophagy [25] . The subunits of Dsl1/ZW10, COG, GARP, and the exocyst complexes share similarities in their α-helical structures [26] . COG's closest relative is the exocyst, which is also octameric and the structure of its subunits SEC3/EXOC1, SEC5/EXOC2, SEC10/EXOC5, SEC15/EXOC6 and EXO70/EXOC7 are similar to COG subunits [27] [28] [29] . MTCs are peripheral membrane proteins that lack membrane anchoring domains and they can only associate with J o u r n a l P r e -p r o o f Journal Pre-proof membranes indirectly. So far, the GARP complex, Dsl1/ZW10 and the exocyst have been shown to be membrane associated via their interactions with Rab GTPases, SNAREs, and/or membrane lipids respectively [30] [31] [32] . The COG complex has eight subunits ( Figure 2 ), encoded by eight different genes, and exists as a hetero-octameric complex or as lobe A (COG1, COG2, COG3, COG4) and lobe B subcomplexes (COG5, COG6, COG7, COG8) [33] . Interestingly, four subunits, COG3, COG4, COG6, and COG8, are more evolutionarily conserved, as are the other four subunits. COG1-COG8 interaction bridges the two subcomplexes together [34] [35] [36] . Mutations that disrupt COG1/8 interactions are detrimental for COG complex functions [37, 38] . The two subcomplexes are observed as lobes in deep-etch electron microscopy (EM) images of the purified COG complex from the bovine brain [39] . Lobe A predominantly associates with the Golgi membrane and can be found on every cisternae [40] , while lobe B preferentially interacts with vesicular membranes [38] . The observed cytoplasmic pool of COG is mainly octameric [29, 36, 38, 39] , while membrane-bound COG can be an octamer or a subcomplex [38] . COG subunits are possibly always bound to the membranes via weak protein-protein interactions with different peripheral or integral membrane proteins. Disruption of these interactions during cell lysis releases COG from the membranes and the soluble pool is likely to be an experimental artifact resulting from the cell lysis procedure. The different arrangements of the COG complex are tied to its functional state, which is described in the 'Current models for COG tethering' section. A slow recovery after photobleaching in Fluorescence Recovery After Photobleaching (FRAP) experiments show that the on/off cycling of COG subunits from the Golgi membrane is very slow [41] , and artificially membrane-glued COG is fully functional [42] . Prolonged depletion or complete knockout of individual COG subunits causes morphological changes in the Golgi (severe fragmentation and dilation of all Golgi cisternae) ( Figure 3a ) that further alters glycosylation [48, 59] , and also affects the endolysosomal system (accumulation of Enlarged Endo-Lysosomal Structures, EELSs) [63] (Figure 3b ), delays retrograde protein trafficking [64] , causes protein missorting and changes the secretion profile [65] . Formation of intra-Golgi SNAREs complex STX5/GOSR1/BET1L/YKT6 [50] and trans-Golgi SNARE complex STX16/STX6/VTI1A/VAMP4 [66] is reduced in COG deficient cells. Another COGrelated trafficking abnormality is the delayed reaction to the fungal metabolite Brefeldin A (BFA) [60, 67] . BFA inhibits the exchange factor GBF1 for small GTPase Arf1, causing COPI coat dysfunction and an artificial fusion of Golgi and ER membranes [68] . The kinetics of both, BFA-induced Golgi "collapse" and subsequent Golgi restoration after BFA wash-out are severely delayed in COG deficient cells. This alteration is often interpreted as COG's requirement for Golgi-ER anterograde and retrograde trafficking [69, 70] . We believe that this interpretation is erroneous and that the altered response to BFA treatment is a secondary manifestation of GBF1, COPI, and/or SNARE malfunction in COG deficient cells [67] . Importantly, many types of COG depleted mammalian cells have wild-type growth and division [48] , emphasizing that the COG complex is generally dispensable for intracellular anterograde trafficking. The deletion of any of the COG subunits in human cells renders the entire complex nonfunctional and results in the degradation of several Golgi resident proteins. Proteins whose stability and function depend on the COG complex are termed COG sensitive proteins, or GEARS [71] . These include many Golgi glycosylation enzymes (MGAT1, MAN2A, B4GALT1, ST6GAL1) [60, 61] and certain members of the trafficking machinery (GS15/BET1L, GS28/GORS2, giantin/GOLGB1, golgin-84/GOLGA5) [71] . Importantly, not every Golgi protein is COG-sensitive; for instance, both the amount and localization of the SNARE STX5, CCT GM130/GOLGA2 and many of the Golgi Rabs are normal in COG-depleted cells [48, 71] (and our unpublished observations). This indicates the existence of COG-independent Golgi recycling/retention mechanism. Analysis of cell lines partially (siRNA) or completely (COG KOs) depleted for COG subunits laid a foundation for the analysis of COG complex deficiencies in humans, described in section 3. J o u r n a l P r e -p r o o f Another important evidence supporting the central role of the COG complex in cellular physiology comes from pathogen-host interaction research. Many intracellular pathogens hijack the host's membrane trafficking machinery to survive within them. Importantly, the COG complex is manipulated by viruses (SARS-Cov-2 [72] , Rift Valley fever [73] , HIV [74] , and Orthopoxvirus [75] ), pathogenic bacteria (Chlamydia [76] and Brucella [77] ), and toxins (SubAB [48, 64] , Shiga [58] , and Cholera [48] ). It is not clear how these diverse groups of pathogens have evolved to rely on COG function. The exact mechanism of COG complex function is still a mystery. Over the years, several models were put forward to explain various mutant phenotypes observed in COG-depleted cells (for review see [33] ). According to the most recent "assembly/disassembly" model [38] , lobe A The assembly/disassembly model would predict that the majority of Golgi associated octameric COG is inert. This then poses the question of what dissociates the two lobes to make them available for another round of vesicle tethering and fusion. While the exact mechanism of COG "recycling" is not understood, one possibility is that the specific binding to vesicular factors (coat, Rabs, SNAREs, CCTs) causes COG disassembly and association of lobe B with the vesicle membrane. Since the past two decades, over a hundred individuals with COG mutations have been identified (see Figure 2 for the location of known mutations in COG subunits). As mentioned in the earlier section, the fidelity of glycosylation is largely dependent on the proper localization of the glycosylation machinery. So, it is not surprising that altered intra-Golgi trafficking due to COG malfunction leads to glycosylation defects [81] . [84] [85] [86] . COG mutations cause CDG-II. In COG-CDGs, the IEF pattern of serum transferrin indicates hyposialylated species, while IEF of apoC-III show increased non-glycosylated apoC-III and decreased monosialo glycoforms [81] . Galactosylation and sialylation defects are also evident by mass-spectrometry in COG-CDGs. Following diagnosis, genotyping is usually done to identify CDG-causing mutations. The next section summarizes the clinical findings of COG-CDG patients and tries to reconcile these findings with current knowledge on the COG complex's cellular functions. (p.L773R) in the COG4 gene [92] . The c.697G>T transcript underwent nonsense-mediated decay. COG4 protein was depleted by 70%, but, curiously, no other subunits were significantly reduced. Glycan mass-spectrometry showed protein sialylation and galactosylation defects. COG4 deficiency hampered COG's function and the Golgi failed to relocate to the ER upon BFA treatment. P2 carried a C>T point mutation at position 2185 in the COG4 gene resulting in R729W change. Though the child was born to unrelated parents -a father heterozygous for the aforementioned mutation and a normal mother, the child seemed to be homozygous for the C>T point mutation. This led the authors to speculate that the maternal allele, which was supposed to be normal, underwent deletion between intron 2 and exon 5. At the molecular level, only 20% of the COG4 protein is expressed. The deficiency of the COG4 subunit affected the stability of all the remaining lobe A subunits and COG5. Sequence and crystal structure analysis of COG4's Cterminal fragment by Richrdson et al [45] found that the mutated arginine is highly conserved. [112] . The cellular phenotype and clinical presentations of COG-CDGs are heterogeneous, but the patients mainly exhibit neurological, morphological, skeletal, and hepatic abnormalities [94, 102, 114] (Figure 4) . One of the main reasons for heterogeneity could be because the mutations have varying effects on both stability and specific function of the mutant COG subunits (see Table 1 ) and the COG complex as a whole. Defects in Golgi glycosylation, particularly reduced sialylation and galactosylation, is the most common feature between COG-CDG patients and receptor binding and uptake [115] . Severe changes in copper homeostasis cause neurodegeneration and COG KO cells show altered stability and localization of copper transporters ATP7A and SLC31A1 [116] . Furthermore, altered glycosphingolipids synthesis in the COG-CDGs could be another possible explanation for neuronal involvement [117] . CDGs associated with the other lobe B subunit, COG5 [102] . Zeevaert et al [114] observed that COG8 deficiency mostly had neurological involvement, whereas growth retardation was a predominant feature in COG1 and COG7-CDG patients and dysmorphic features of face and hands are common in all CDG patients. Given the small cohort of COG-CDGs that have been reported, it is difficult to make further correlations between the affected subunit and clinical manifestations. Comparisons between the clinical phenotype of COG-CDGs should take into consideration the effect of the mutation on the affected COG subunit. In the recent retrospective analysis of COG-CDGs, Haijes et al [118] made such a comparison and concluded that lobe A mutations are more detrimental than lobe B mutation, but the discussion is not settled mainly because of the small number of known mutations and lack of robust data on these patients. Treating congenital multi-systemic disorders is challenging and one single approach could be insufficient. A good example is the treatment of MPI-CDG. Clinical presentation of MPI-CDG includes but is not limited to liver fibrosis and gastrointestinal complications, thrombosis, and hypoglycemia. With oral mannose intake and liver transplantation, the patient improved dramatically [119] . Similarly, galactose supplementation has been used to alleviate some of the pathologies of PGM1-CDG [120] . Patients with mutations in TMEM165 have CDG-II and their J o u r n a l P r e -p r o o f Journal Pre-proof glycan profiles reveal hypogalactosylation. Two patients with TMEM165-CDG responded positively to galactose supplementation [121] . COG mutations cause a wide range of defects that directly (glycosylation enzymes, sugar transporters) and indirectly (Golgi homeostasis, cisternal compartmentalization, and stability of SNAREs and GEARs) impact glycosylation and other trafficking processes. Interestingly, data from our lab indicates that TMEM165 is dependent on the COG complex for its stability and its depletion in COG KO cells may contribute to misglycosylation (unpublished observations). If this prediction is correct, galactose therapy may partially alleviate hypoglycosylation in COG-CDGs. It is important to note that COG mutations not only affect the Golgi, but also the endolysosomal system, causing altered secretion and potential malfunction of the lysosomal degradation machinery [65] ; therefore, treatment of COG-CDG patients should combine strategies used for the treatment of both CDG and lysosomal disorders. Antisense therapy and gene therapy have not yet been well fully explored for the treatment of CDGs [122] . In TMEM165-CDG patient fibroblast, antisense therapy successfully rescued exon skipping due to an intronic mutation and drove expression of the WT protein [123] . This approach is applicable where an intronic mutation leads to the activation of a cryptic splice site upstream of the canonical splice site. Antisense morpholino oligonucleotides block the cryptic splice site and promote canonical splicing. With the advancement in research on gene therapy, it could soon become a reality for the treatment of COG-CDGs. Theoretically, gene therapy should have promising outcomes in the treatment of COG-CDGs since the introduction of the WT copy of the affected COG subunit fully rescues COG-CDG defects in cell lines [63] . At present, the significant challenges/risks include safe vehicular delivery and targeted integration of the delivered gene. Gene therapy, through intravenous and intramuscular liposomal delivery, rescued GNE-CDG (UDP-GlcNAc2 epimerase-CDG) in mice [124] . The establishment of a reliable animal COG disease model will benefit the development of therapeutics. There are some encouraging attempts to explore fly [125] and fish [91] Nevertheless, the molecular mechanism of COG-mediated vesicle tethering is only speculative and the exact molecular reactions and sequence of events leading to docking and fusion of retrograde membrane carriers at the Golgi are not fully understood. COG has multiple interactions with other components of a vesicle fusion machinery, but the temporal sequence of these protein-protein interactions is not clear. The prediction is that at least some COG interactions with the Golgi trafficking machinery are sequential rather than simultaneous since individual subunits interact with more than one trafficking partner. In vitro reconstitution of the COG complex function will undoubtedly help in answering this and other mechanistic questions. So far, successful in vitro reconstitution was achieved only for one MTC, yeast HOPS complex [126] . Which molecules are responsible for the COG complex's recruitment to the Golgi membrane? CATCHRs employ diverse strategies for membrane association: Dsl1 is membrane-associated by ER anchored SNAREs [127] . The GARP complex is recruited by Arl5 [128] In yeast, the cytoplasm to vacuole targeting (Cvt) pathway is dependent on the activity of COG lobe A subunits. Autophagosome formation and the sorting of Atg proteins to the site of nucleation is affected in COG mutant cells [132] . This suggests that the COG complex might be involved in tethering events leading to autophagosome formation. Furthermore, one of the Atg proteins, Atg6 is dispersed in COG2 yeast mutants while several other Atg proteins have been shown to interact with COG subunits in yeast two-hybrid assays. At the TGN, COG8 controls Atg9 trafficking in collaboration with Arl1 and Al3 GTPases [133] . If indeed COG is involved in autophagy, it would be worth studying the status of autophagy in COG-CDG patients and if found faulty, weigh in its impact on the COG-CDG phenotype. The first COG subunits were discovered by the Monty Krieger lab 35 years ago [56] ; it took almost 20 years to realize that these proteins act together as an evolutionarily conserved vesicle tethering complex [29, 39] and that mutations in COG subunits are responsible for a whole family of human CDG-II diseases [105, 114] . We hope that the potent combination of molecular We J o u r n a l P r e -p r o o f A prize for membrane magic Evolutionary origins and specialisation of membrane transport The mechanisms of vesicle budding and fusion The Evolution of Organellar Coat Complexes and Organization of the Eukaryotic Cell Assembly of COPI and COPII Vesicular Coat Proteins on Membranes, Annual review of biophysics Motor-cargo adaptors at the organelle-cytoskeleton interface Rab GTPases and their interacting protein partners: Structural insights into Rab functional diversity, Small Gtpases ER to Golgi-Dependent Protein Secretion: The Conventional Pathway Transport carrier tethering -how vesicles are captured by organelles Mechanism of membrane fusion: protein-protein interaction and beyond Hypothesis -buttressed rings assemble, clamp, and release SNAREpins for synaptic transmission Breaking the COPI monopoly on Golgi recycling Maturation-driven transport and AP-1-dependent recycling of a secretory cargo in the Golgi Recycling of Golgi glycosyltransferases requires direct binding to coatomer Golgi phosphoprotein 3 triggers signal-mediated incorporation of glycosyltransferases into coatomer-coated (COPI) vesicles Sorting of membrane proteins in the secretory pathway The organizing potential of sphingolipids in intracellular membrane transport A tale of short tails, through thick and thin: investigating the sorting mechanisms of Golgi enzymes Mechanisms of protein retention in the Golgi Neutralization of pH in the Golgi apparatus causes redistribution of glycosyltransferases and changes in the O-glycosylation of mucins Functional organization of Golgi N-and O-glycosylation pathways involves pHdependent complex formation that is impaired in cancer cells A Golgi-associated redox switch regulates catalytic activation and cooperative functioning of ST6Gal-I with B4GalT-I, Redox biology Glycosylation Quality Control by the Golgi Structure Structure of membrane tethers and their role in fusion Tethering Factors as Organizers of Intracellular Vesicular Traffic The structures of exocyst subunit Exo70p and the Exo84p C-terminal domains reveal a common motif The structure of the exocyst subunit Sec6p defines a conserved architecture with diverse roles The Sec34/35 Golgi transport complex is related to the exocyst, defining a family of complexes involved in multiple steps of membrane traffic Membrane association and functional regulation of Sec3 by phospholipids and Cdc42 Exo70 interacts with phospholipids and mediates the targeting of the exocyst to the plasma membrane Membrane targeting of the yeast exocyst complex Maintaining order: COG complex controls Golgi trafficking, processing, and sorting Subunit architecture of the conserved oligomeric Golgi complex Genetic analysis of the subunit organization and function of the conserved oligomeric golgi (COG) complex: studies of COG5-and COG7-deficient mammalian cells Cog1p plays a central role in the organization of the yeast conserved oligomeric Golgi complex A new inborn error of glycosylation due to a Cog8 deficiency reveals a critical role for the Cog1-Cog8 interaction in COG complex formation COG lobe B sub-complex engages v-SNARE GS15 and functions via regulated interaction with lobe A sub-complex Characterization of a mammalian Golgi-localized protein complex, COG, that is required for normal Golgi morphology and function IntraGolgi distribution of the Conserved Oligomeric Golgi (COG) complex Multipronged interaction of the COG complex with intracellular membranes Membrane detachment is not essential for COG complex function Structural analysis of conserved oligomeric Golgi complex subunit 2 Cog5-Cog7 crystal structure reveals interactions essential for the function of a multisubunit tethering complex Structural basis for a human glycosylation disorder caused by mutation of the COG4 gene Molecular architecture of the complete COG tethering complex The In Vivo Architecture of the Exocyst Provides Structural Basis for Exocytosis COG Complex Complexities: Detailed Characterization of a Complete Set of HEK293T Cells Lacking Individual COG Subunits The Golgi puppet master: COG complex at center stage of membrane trafficking interactions Interaction of the conserved oligomeric Golgi complex with t-SNARE Syntaxin5a/Sed5 enhances intra-Golgi SNARE complex stability Direct interaction between the COG complex and the SM protein, Sly1, is required for Golgi SNARE pairing The interaction of two tethering factors, p115 and COG complex, is required for Golgi integrity Interaction of Golgin-84 with the COG complex mediates the intra-Golgi retrograde transport New mutants of Saccharomyces cerevisiae affected in the transport of proteins from the endoplasmic reticulum to the Golgi complex The Sec34/Sec35p complex, a Ypt1p effector required for retrograde intra-Golgi trafficking, interacts with Golgi SNAREs and COPI vesicle coat proteins Three types of low density lipoprotein receptor-deficient mutant have pleiotropic defects in the synthesis of N-linked, Olinked, and lipid-linked carbohydrate chains Purification and characterization of a novel 13 S hetero-oligomeric protein complex that stimulates in vitro Golgi transport Cog3p depletion blocks vesicle-mediated Golgi retrograde trafficking in HeLa cells COG complex-mediated recycling of Golgi glycosyltransferases is essential for normal protein glycosylation Conserved oligomeric Golgi complex specifically regulates the maintenance of Golgi glycosylation machinery Fluorescent microscopy as a tool to elucidate dysfunction and mislocalization of Golgi glycosyltransferases in COG complex depleted mammalian cells Differential effects of lobe A and lobe B of the Conserved Oligomeric Golgi complex on the stability of {beta}1,4-galactosyltransferase 1 and {alpha}2,6-sialyltransferase 1 Defects in COG-Mediated Golgi Trafficking Alter Endo-Lysosomal System in Human Cells The COG Complex, Rab6 and COPI Define a Novel Golgi Retrograde Trafficking Pathway that is Exploited by SubAB Toxin More than just sugars: Conserved oligomeric Golgi complex deficiency causes glycosylation-independent cellular defects The COG complex interacts directly with Syntaxin 6 and positively regulates endosome-to-TGN retrograde transport Mislocalization of large ARF-GEFs as a potential mechanism for BFA resistance in COGdeficient cells Dynamics of GBF1, a Brefeldin A-sensitive Arf1 exchange factor at the Golgi COG5-CDG: expanding the clinical spectrum Fatal outcome due to deficiency of subunit 6 of the conserved oligomeric Golgi complex leading to a new type of congenital disorders of glycosylation The COG and COPI complexes interact to control the abundance of GEARs, a subset of Golgi integral membrane proteins Multi-level proteomics reveals host-perturbation strategies of SARS-CoV-2 and SARS-CoV, in, bioRxiv A Haploid Genetic Screen Identifies Heparan Sulfate Proteoglycans Supporting Rift Valley Fever Virus Infection Target silencing of components of the conserved oligomeric Golgi complex impairs HIV-1 replication Conserved Oligomeric Golgi (COG) Complex Proteins Facilitate Orthopoxvirus Entry Chlamydia trachomatis hijacks intra-Golgi COG complex-dependent vesicle trafficking pathway A Brucella Type IV Effector Targets the COG Tethering Complex to Remodel Host Secretory Traffic and Promote Intracellular Replication Ral GTPases regulate exocyst assembly through dual subunit interactions Exocyst dynamics during vesicle tethering and fusion Exocyst structural changes associated with activation of tethering downstream of Rho/Cdc42 GTPases COG defects, birth and rise! Congenital disorders of glycosylation (CDG): Quo vadis? Congenital disorders of glycosylation How to find and diagnose a CDG due to defective Nglycosylation Plasma N-glycan profiling by mass spectrometry for congenital disorders of glycosylation type II Patients with unsolved congenital disorders of glycosylation type II can be subdivided in six distinct biochemical groups Conserved oligomeric Golgi complex subunit 1 deficiency reveals a previously uncharacterized congenital disorder of glycosylation type II Cerebrocostomandibular-like syndrome and a mutation in the conserved oligomeric Golgi complex, subunit 1 oligomeric golgi complex cause a congenital disorder of glycosylation Golgi function and dysfunction in the first COG4-deficient CDG type II patient A Recurrent De Novo Heterozygous COG4 Substitution Leads to Saul-Wilson Syndrome, Disrupted Vesicular Trafficking, and Altered Proteoglycan Glycosylation Identification of the first COG-CDG patient of Indian origin Deficiency in COG5 causes a moderate form of congenital disorders of glycosylation COG5-CDG with a Mild Neurohepatic Presentation A Mild Form of COG5 Defect Showing Early-Childhood-Onset Friedreich's-Ataxia-Like Phenotypes with Isolated Cerebellar Atrophy Novel compound heterozygous COG5 mutations in a Chinese male patient with severe clinical symptoms and type IIi congenital disorder of glycosylation: A case report, Experimental and therapeutic medicine Identification of Two Novel Mutations in COG5 Causing Congenital Disorder of Glycosylation MALDI-MS profiling of serum O-glycosylation and N-glycosylation in COG5-CDG Deficiency of Subunit 6 of the Conserved Oligomeric Golgi Complex (COG6-CDG): Second Patient, Different Phenotype A novel syndrome of hypohidrosis and intellectual disability is linked to COG6 deficiency Secondary Hemophagocytic Syndrome Associated with COG6 Gene Defect: Report and Review Key features and clinical variability of COG6-CDG Compound heterozygous variants of the COG6 gene in a Chinese patient with deficiency of subunit 6 of the conserved oligomeric Golgi complex (COG6-CDG) Al-Kouatly, Nonimmune hydrops fetalis and congenital disorders of glycosylation: A systematic literature review Mutation of the COG complex subunit gene COG7 causes a lethal congenital disorder Clinical and biochemical presentation of siblings with COG-7 deficiency, a lethal multiple O-and N-glycosylation disorder A common mutation in the COG7 gene with a consistent phenotype including microcephaly, adducted thumbs, growth retardation, VSD and episodes of hyperthermia COG-7-deficient Human Fibroblasts Exhibit Altered Recycling of Golgi Proteins A new mutation in COG7 extends the spectrum of COG subunit deficiencies COG8 deficiency causes new congenital disorder of glycosylation type IIh Further delineation of COG8-CDG: A case with novel compound heterozygous mutations diagnosed by targeted exome sequencing The first case of antenatal presentation in COG8-congenital disorder of glycosylation with a novel splice site mutation and an extended phenotype Deficiency of the Cog8 subunit in normal and CDG-derived cells impairs the assembly of the COG and Golgi SNARE complexes Deficiencies in subunits of the Conserved Oligomeric Golgi (COG) complex define a novel group of Congenital Disorders of Glycosylation N-glycosylation in regulation of the nervous system The interactome of the copper transporter ATP7A Conserved Oligomeric Golgi and Neuronal Vesicular Trafficking Hypothesis: lobe A (COG1-4)-CDG causes a more severe phenotype than lobe B (COG5-8)-CDG Successful liver transplantation and long-term follow-up in a patient with MPI-CDG Oral D-galactose supplementation in PGM1-CDG Galactose Supplementation in Patients With TMEM165-CDG Rescues the Glycosylation Defects From Bench to Bedside Antisense-mediated therapeutic pseudoexon skipping in TMEM165-CDG Safety and in vivo expression of a GNE-transgene: a novel treatment approach for hereditary inclusion body myopathy-2 COG7 deficiency in Drosophila generates multifaceted developmental, behavioral and protein glycosylation phenotypes HOPS recognizes each SNARE, assembling ternary trans-complexes for rapid fusion upon engagement with the 4th SNARE, eLife Structural basis for the binding of SNAREs to the multisubunit tethering complex Dsl1 ARFRP1 functions upstream of ARL1 and ARL5 to coordinate recruitment of distinct tethering factors to the trans-Golgi network Structure and mechanism in membrane trafficking A new role for RINT-1 in SNARE complex assembly at the trans-Golgi network in coordination with the COG complex COG6 interacts with a subset of the Golgi SNAREs and is important for the Golgi complex integrity The conserved oligomeric Golgi complex is involved in double-membrane vesicle formation during autophagy The Arl3 and Arl1 GTPases co-operate with Cog8 to regulate selective autophagy via Atg9 trafficking Using medical exome sequencing to identify the causes of neurodevelopmental disorders: Experience of 2 clinical units and 216 patients Genetic polymorphisms of hspa1b and hspa1l in infertile men