Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Gege Xu is active.

Publication


Featured researches published by Gege Xu.


Cell Host & Microbe | 2016

Depletion of Butyrate-Producing Clostridia from the Gut Microbiota Drives an Aerobic Luminal Expansion of Salmonella

Fabian Rivera-Chávez; Lillian F. Zhang; Franziska Faber; Christopher A. Lopez; Mariana X. Byndloss; Erin E. Olsan; Gege Xu; Eric M. Velazquez; Carlito B. Lebrilla; Sebastian E. Winter; Andreas J. Bäumler

The mammalian intestine is host to a microbial community that prevents pathogen expansion through unknown mechanisms, while antibiotic treatment can increase susceptibility to enteric pathogens. Here we show that streptomycin treatment depleted commensal, butyrate-producing Clostridia from the mouse intestinal lumen, leading to decreased butyrate levels, increased epithelial oxygenation, and aerobic expansion of Salmonella enterica serovar Typhimurium. Epithelial hypoxia and Salmonella restriction could be restored by tributyrin treatment. Clostridia depletion and aerobic Salmonella expansion were also observed in the absence of streptomycin treatment in genetically resistant mice but proceeded with slower kinetics and required the presence of functional Salmonella type III secretion systems. The Salmonella cytochrome bd-II oxidase synergized with nitrate reductases to drive luminal expansion, and both were required for fecal-oral transmission. We conclude that Salmonella virulence factors and antibiotic treatment promote pathogen expansion through the same mechanism: depletion of butyrate-producing Clostridia to elevate epithelial oxygenation, allowing aerobic Salmonella growth.


Science | 2017

Microbiota-activated PPAR-γ signaling inhibits dysbiotic Enterobacteriaceae expansion

Mariana X. Byndloss; Erin E. Olsan; Fabian Rivera-Chávez; Connor R. Tiffany; Stephanie A. Cevallos; Kristen L. Lokken; Teresa P. Torres; Austin J. Byndloss; Franziska Faber; Yandong Gao; Yael Litvak; Christopher A. Lopez; Gege Xu; Eleonora Napoli; Cecilia Giulivi; Renée M. Tsolis; Alexander Revzin; Carlito B. Lebrilla; Andreas J. Bäumler

Healthy guts exclude oxygen Normally, the lumen of the colon lacks oxygen. Fastidiously anaerobic butyrate-producing bacteria thrive in the colon; by ablating these organisms, antibiotic treatment removes butyrate. Byndloss et al. discovered that loss of butyrate deranges metabolic signaling in gut cells (see the Perspective by Cani). This induces nitric oxidase to generate nitrate in the lumen and disables β-oxidation in epithelial cells that would otherwise mop up stray oxygen before it enters the colon. Simultaneously, regulatory T cells retreat, and inflammation is unchecked, which contributes yet more oxygen species to the colon. Then, facultative aerobic pathogens, such as Escherichia coli and Salmonella enterica, can take advantage of the altered environment and outgrow any antibiotic-crippled and benign anaerobes. Science, this issue p. 570; see also p. 548 Butyrate-producing microbes contribute to synergism between epithelial cell metabolism and immune response regulation to maintain gut heath. Perturbation of the gut-associated microbial community may underlie many human illnesses, but the mechanisms that maintain homeostasis are poorly understood. We found that the depletion of butyrate-producing microbes by antibiotic treatment reduced epithelial signaling through the intracellular butyrate sensor peroxisome proliferator–activated receptor γ (PPAR-γ). Nitrate levels increased in the colonic lumen because epithelial expression of Nos2, the gene encoding inducible nitric oxide synthase, was elevated in the absence of PPAR-γ signaling. Microbiota-induced PPAR-γ signaling also limits the luminal bioavailability of oxygen by driving the energy metabolism of colonic epithelial cells (colonocytes) toward β-oxidation. Therefore, microbiota-activated PPAR-γ signaling is a homeostatic pathway that prevents a dysbiotic expansion of potentially pathogenic Escherichia and Salmonella by reducing the bioavailability of respiratory electron acceptors to Enterobacteriaceae in the lumen of the colon.


mSphere | 2017

Persistence of Supplemented Bifidobacterium longum subsp. infantis EVC001 in Breastfed Infants

Steven A. Frese; Andra A. Hutton; Lindsey N. Contreras; Claire A. Shaw; Michelle C. Palumbo; Giorgio Casaburi; Gege Xu; Jasmine C.C. Davis; Carlito B. Lebrilla; Bethany M. Henrick; Samara L. Freeman; Daniela Barile; J. Bruce German; David A. Mills; Jennifer T. Smilowitz; Mark A. Underwood

The gut microbiome in early life plays an important role for long-term health and is shaped in large part by diet. Probiotics may contribute to improvements in health, but they have not been shown to alter the community composition of the gut microbiome. Here, we found that breastfed infants could be stably colonized at high levels by provision of B. infantis EVC001, with significant changes to the overall microbiome composition persisting more than a month later, whether the infants were born vaginally or by caesarean section. This observation is consistent with previous studies demonstrating the capacity of this subspecies to utilize human milk glycans as a nutrient and underscores the importance of pairing a probiotic organism with a specific substrate. Colonization by B. infantis EVC001 resulted in significant changes to fecal microbiome composition and was associated with improvements in fecal biochemistry. The combination of human milk and an infant-associated Bifidobacterium sp. shows, for the first time, that durable changes to the human gut microbiome are possible and are associated with improved gut function. ABSTRACT Attempts to alter intestinal dysbiosis via administration of probiotics have consistently shown that colonization with the administered microbes is transient. This study sought to determine whether provision of an initial course of Bifidobacterium longum subsp. infantis (B. infantis) would lead to persistent colonization of the probiotic organism in breastfed infants. Mothers intending to breastfeed were recruited and provided with lactation support. One group of mothers fed B. infantis EVC001 to their infants from day 7 to day 28 of life (n = 34), and the second group did not administer any probiotic (n = 32). Fecal samples were collected during the first 60 postnatal days in both groups. Fecal samples were assessed by 16S rRNA gene sequencing, quantitative PCR, mass spectrometry, and endotoxin measurement. B. infantis-fed infants had significantly higher populations of fecal Bifidobacteriaceae, in particular B. infantis, while EVC001 was fed, and this difference persisted more than 30 days after EVC001 supplementation ceased. Fecal milk oligosaccharides were significantly lower in B. infantis EVC001-fed infants, demonstrating higher consumption of human milk oligosaccharides by B. infantis EVC001. Concentrations of acetate and lactate were significantly higher and fecal pH was significantly lower in infants fed EVC001, demonstrating alterations in intestinal fermentation. Infants colonized by Bifidobacteriaceae at high levels had 4-fold-lower fecal endotoxin levels, consistent with observed lower levels of Gram-negative Proteobacteria and Bacteroidetes. IMPORTANCE The gut microbiome in early life plays an important role for long-term health and is shaped in large part by diet. Probiotics may contribute to improvements in health, but they have not been shown to alter the community composition of the gut microbiome. Here, we found that breastfed infants could be stably colonized at high levels by provision of B. infantis EVC001, with significant changes to the overall microbiome composition persisting more than a month later, whether the infants were born vaginally or by caesarean section. This observation is consistent with previous studies demonstrating the capacity of this subspecies to utilize human milk glycans as a nutrient and underscores the importance of pairing a probiotic organism with a specific substrate. Colonization by B. infantis EVC001 resulted in significant changes to fecal microbiome composition and was associated with improvements in fecal biochemistry. The combination of human milk and an infant-associated Bifidobacterium sp. shows, for the first time, that durable changes to the human gut microbiome are possible and are associated with improved gut function.


Journal of Human Lactation | 2018

Composition and Variation of Macronutrients, Immune Proteins, and Human Milk Oligosaccharides in Human Milk From Nonprofit and Commercial Milk Banks

Laura Meredith-Dennis; Gege Xu; Elisha Goonatilleke; Carlito B. Lebrilla; Mark A. Underwood; Jennifer T. Smilowitz

Background: When human milk is unavailable, banked milk is recommended for feeding premature infants. Milk banks use processes to eliminate pathogens; however, variability among methods exists. Research aim: The aim of this study was to compare the macronutrient (protein, carbohydrate, fat, energy), immune-protective protein, and human milk oligosaccharide (HMO) content of human milk from three independent milk banks that use pasteurization (Holder vs. vat techniques) or retort sterilization. Methods: Randomly acquired human milk samples from three different milk banks (n = 3 from each bank) were analyzed for macronutrient concentrations using a Fourier transform mid-infrared spectroscopy human milk analyzer. The concentrations of IgA, IgM, IgG, lactoferrin, lysozyme, α-lactalbumin, α antitrypsin, casein, and HMO were analyzed by mass spectrometry. Results: The concentrations of protein and fat were significantly (p < .05) less in the retort sterilized compared with the Holder and vat pasteurized samples, respectively. The concentrations of all immune-modulating proteins were significantly (p < .05) less in the retort sterilized samples compared with vat and/or Holder pasteurized samples. The total HMO concentration and HMOs containing fucose, sialic acid, and nonfucosylated neutral sugars were significantly (p < .05) less in retort sterilized compared with Holder pasteurized samples. Conclusion: Random milk samples that had undergone retort sterilization had significantly less immune-protective proteins and total and specific HMOs compared with samples that had undergone Holder and vat pasteurization. These data suggest that further analysis of the effect of retort sterilization on human milk components is needed prior to widespread adoption of this process.


Chemical Reviews | 2018

Mass Spectrometry Approaches to Glycomic and Glycoproteomic Analyses

L. Renee Ruhaak; Gege Xu; Qiongyu Li; Elisha Goonatilleke; Carlito B. Lebrilla

Glycomic and glycoproteomic analyses involve the characterization of oligosaccharides (glycans) conjugated to proteins. Glycans are produced through a complicated nontemplate driven process involving the competition of enzymes that extend the nascent chain. The large diversity of structures, the variations in polarity of the individual saccharide residues, and the poor ionization efficiencies of glycans all conspire to make the analysis arguably much more difficult than any other biopolymer. Furthermore, the large number of glycoforms associated with a specific protein site makes it more difficult to characterize than any post-translational modification. Nonetheless, there have been significant progress, and advanced separation and mass spectrometry methods have been at its center and the main reason for the progress. While glycomic and glycoproteomic analyses are still typically available only through highly specialized laboratories, new software and workflow is making it more accessible. This review focuses on the role of mass spectrometry and separation methods in advancing glycomic and glycoproteomic analyses. It describes the current state of the field and progress toward making it more available to the larger scientific community.


Analytical Chemistry | 2018

Recent Advances in the Mass Spectrometry Methods for Glycomics and Cancer

Muchena J. Kailemia; Gege Xu; Maurice Wong; Qiongyu Li; Elisha Goonatilleke; Frank Leon; Carlito B. Lebrilla

The review focuses on recent aspects (last three years) of glycosylation analyses that provide relevant information about cancer. It includes recent development in glycan and protein enrichment methods for discovery of cancer markers. It will however focus on the recent technological developments in mass spectrometry (MS), bioinformatics and separation methods as they apply toward identifying cancer markers. More specifically, it will cover advances in matrix-assisted laser desorption/ionization (MALDI), electrospray ionization (ESI), capillary electrophoresis (CE), and liquid chromatography (LC) coupled to mass spectrometry. The discussions will include glycans, recently identified as potential markers for cancer that have been discovered using the highlighted technologies. We will also discuss emerging glycoproteomic techniques and site-specific methods, and how these methods are being utilized for cancer biomarker discovery. The large amount of data and the complexity of glycoproteomic analysis have been the impetus for developing bioinformatic methods for assigning glycosylation sites and characterizing the potentially very large site- or microheterogeneity. This review will cover the most recent advancements in biomarker discovery of N- and O-glycosylation of proteins as well as the glycolipids. This group collectively constitutes glycosylation on the cell membrane or the glycocalyx. The review will also highlight methods that are highly reproducible, with low coefficient of variation (CV), and scalable for large sample sets. The reader is also referred to other notable earlier reviews on glycomic biomarkers for cancer. Mereiter et al. describe the recent glycomic effort in gastrointestinal cancer.1 A review focused on N-glycomic analysis of colorectal cancer has been published by Sethi and Fanayan.2 N-Glycan, O-glycan, and glycolipid characteristics of colorectal cancer were reviewed by Holst et al.3 Muchena et al. have provided a more general review of glycan biomarkers covering up to the current review period.4 The field of glycoscience also covers a broad area of structures and may include highly anionic (glycosaminoglycans) and monosaccharide (e.g. O-GlcNAc) modifications that require their specific and unique sets of analytical tools. The latter topics are not covered in this review. 1.1. Background of Glycosylation and Cancer There is nearly 50 years of research illustrating that changes in glycosylation accompany cancer.5 Glycosylation is a dynamic process intimately involved in key processes in cells, including cell-cell and cell-extracellular communication as well as cell-cell adhesion, and cellular metabolism. Glycans expressed in several types of glycoconjugates are known to change during cancer genesis and progression.6 These changes increase the structural heterogeneity and alter the functions of cells.7 Glycosylation has been found to enable tumor-induced immunomodulation and metastasis.8–10 The cell-surface structures allow the immune cells to differentiate self/normal cells from non-self/abnormal cells.11 For example, terminal residues on N-glycans, such as sialic acids, are involved in immunity and cell-cell communication.12 Changes in glycosylation of adhesion proteins can largely influence their binding properties, leading to altered cell-cell or cell-matrix contacts.13 Other types of glycans are also involved in cancer. Gangliosides and sphingolipids are involved in transmembrane communication vital in tumor cell growth and invasion.14 Glycosaminoglycans are involved in tumor cell migration15 and motility.16–18 The search for effective markers is aided by the understanding of how glycans are synthesized. The glycan biosynthetic process is a non-template process involving multiple enzymes, some performing competing activities. It is estimated that more than 300 metabolic enzymes, composed of glycosyltransferases and glycosidases, are involved in the biosynthesis and processing of glycans.19–20 The best-known series of pathways belongs to the production of N-glycans (Figure 1). They illustrate the large degree of structural heterogeneity in glycosylation. N-Glycans are produced in a step-wise process beginning with the production of high mannose structures on a lipid, which are transferred to the nascent polypeptide chain to guide protein folding. Once folded, the glycans are then trimmed back and extended to form complex and hybrid structures. The folded protein can be secreted with glycans that range from early in the process to yield high mannose structures to later in the process corresponding to complex or hybrid structures. The number of structures for one glycosylation site can vary by a large degree, from a handful for transferrin21 to over 70 structures for IgG, the most abundant serum glycoprotein.22–23 Open in a separate window Figure 1. Representation of the glycosylation pathway of proteins. The pathway illustrates the complexity and heterogeneity of structures. The proteins may exit the pathway with various levels of glycosylation.


Glycobiology | 2017

Enterocyte glycosylation is responsive to changes in extracellular conditions: implications for membrane functions

Dayoung Park; Gege Xu; Mariana Barboza; Ishita M. Shah; Maurice Wong; Helen E. Raybould; David A. Mills; Carlito B. Lebrilla

Epithelial cells in the lining of the intestines play critical roles in maintaining homeostasis while challenged by dynamic and sudden changes in luminal contents. Given the high density of glycosylation that encompasses their extracellular surface, environmental changes may lead to extensive reorganization of membrane-associated glycans. However, neither the molecular details nor the consequences of conditional glycan changes are well understood. Here we assessed the sensitivity of Caco-2 and HT-29 membrane N-glycosylation to variations in (i) dietary elements, (ii) microbial fermentation products and (iii) cell culture parameters relevant to intestinal epithelial cell growth and survival. Based on global LC-MS glycomic and statistical analyses, the resulting glycan expression changes were systematic, dependent upon the conditions of each controlled environment. Exposure to short chain fatty acids produced significant increases in fucosylation while further acidification promoted hypersialylation. Notably, among all conditions, increases of high mannose type glycans were identified as a major response when extracellular fructose, galactose and glutamine were independently elevated. To examine the functional consequences of this discrete shift in the displayed glycome, we applied a chemical inhibitor of the glycan processing mannosidase, globally intensifying high mannose expression. The data reveal that upregulation of high mannose glycosylation has detrimental effects on basic intestinal epithelium functions by altering permeability, host-microbe associations and membrane protein activities.


bioRxiv | 2018

Infection-generated electric field in gut epithelium drives bidirectional migration of macrophages

Yaohui Sun; Brian Reid; Fernando Ferreira; Guillaume Luxardi; Li Ma; Kristen L. Lokken; Kan Zhu; Gege Xu; Yuxin Sun; Volodymyr Ryzhuk; Betty P. Guo; Carlito B. Lebrilla; Emanual Maverakis; Alex Mogilner; Min Zhao

Many bacterial pathogens hijack macrophages to egress from the port of entry to the lymphatic/blood-stream, causing dissemination of life-threatening infections. However, the underlying mechanisms are not well understood. Here, we report that Salmonella infection generates directional electric fields (EF) in the follicle-associated epithelium of mouse cecum. In vitro application of an EF, mimicking the infection-generated electric field (IGEF), induces directional migration of primary mouse macrophages to the anode, which is reversed to the cathode upon Salmonella infection. This infection-dependent directional switch is independent of the Salmonella pathogenicity island 1 (SPI-1) type III secretion system. The switch is accompanied by a reduction of sialic acids on glycosylated surface components during phagocytosis of bacteria, which is absent in macrophages challenged by microspheres. Moreover, enzymatic cleavage of terminally exposed sialic acids reduces macrophage surface negativity and severely impairs directional migration of macrophages in response to EF. Based on these findings, we propose that macrophages are attracted to the site of infection by a combination of chemotaxis and galvanotaxis; after phagocytosis of bacteria, surface electrical properties of the macrophage change, and galvanotaxis directs the cells away from the site of infection. Abbreviations CFU, colony-forming unit; Con A, Concanavalin A; EF, electric field; FAE, follicle-associated epithelium; GNL, Galanthus Nivalis lectin; IGEF, infection-generated electric field; Ji, electric current density; MAL-2, Maackia Amurensis lectin II; MLN, mesenteric lymph node; MOI, multiplicity of infection; nMFI, normalized mean fluorescence intensity; RCA-1, Ricinus Communis Agglutinin I; SNA, Sambucus Nigra lectin; S. Typhimurium, Salmonella enterica serotype Typhimurium; SPI-1, Salmonella pathogenicity island 1; PDMS, polydimethylsiloxane; TEP, trans-epithelial potential difference; TLR, Toll-like receptors; WGEF, wound-generated electric field


Stem cell reports | 2018

FGF2 Induces Migration of Human Bone Marrow Stromal Cells by Increasing Core Fucosylations on N-Glycans of Integrins

Baarkullah Awan; David Turkov; Cameron Schumacher; Antonio Jacobo; Amber McEnerney; Ashley Ramsey; Gege Xu; Dayoung Park; Stefanos Kalomoiris; Wei Yao; Li-En Jao; Miguel L. Allende; Carlito B. Lebrilla; Fernando A. Fierro

Summary Since hundreds of clinical trials are investigating the use of multipotent stromal cells (MSCs) for therapeutic purposes, effective delivery of the cells to target tissues is critical. We have found an unexplored mechanism, by which basic fibroblast growth factor (FGF2) induces expression of fucosyltransferase 8 (FUT8) to increase core fucosylations of N-linked glycans of membrane-associated proteins, including several integrin subunits. Gain- and loss-of-function experiments show that FUT8 is both necessary and sufficient to induce migration of MSCs. Silencing FUT8 also affects migration of MSCs in zebrafish embryos and a murine bone fracture model. Finally, we use in silico modeling to show that core fucosylations restrict the degrees of freedom of glycans on the integrin’s surface, hence stabilizing glycans on a specific position. Altogether, we show a mechanism whereby FGF2 promotes migration of MSCs by modifying N-glycans. This work may help improve delivery of MSCs in therapeutic settings.


Scientific Reports | 2018

Intact glycosphingolipidomic analysis of the cell membrane during differentiation yields extensive glycan and lipid changes

Maurice Wong; Gege Xu; Dayoung Park; Mariana Barboza; Carlito B. Lebrilla

Glycosphingolipids (GSLs) are found in cellular membranes of most organisms and play important roles in cell-cell recognition, signaling, growth, and adhesion, among others. A method based on nanoflow high performance liquid chromatography-chip-quadrupole-time-of-flight mass spectrometry (nanoHPLC Chip-Q-TOF MS) was applied towards identifying and quantifying intact GSLs from a variety of samples, including cultured cell lines and animal tissue. The method provides the composition and sequence of the glycan, as well as variations in the ceramide portion of the GSL. It was used to profile the changes in the glycolipidome of Caco-2 cells as they undergo differentiation. A total of 226 unique GSLs were found among Caco-2 samples from five differentiation time-points. The method provided a comprehensive glycolipidomic profile of a cell during differentiation to yield the dynamic variation of intact GSL structures.

Collaboration


Dive into the Gege Xu's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Maurice Wong

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dayoung Park

University of California

View shared research outputs
Top Co-Authors

Avatar

Erin E. Olsan

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge